Biochemistry Lectures

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 139

0

STATE UNIVERSITY OF MEDICINE AND PHARMACY NI COLAE TESTEMI TANU


BIOCHEMISTRY AND CLINICAL BIOCHEMISTRY DEPARTMENT



LUDMILA GAVRILIUC





BIOCHEMISTRY
Lectures for students of Medical Departments









1
CHISINAU
2011
STATE UNIVERSITY OF MEDICINE AND PHARMACY NI COLAE TESTEMI TANU
BIOCHEMISTRY AND CLINICAL BIOCHEMISTRY DEPARTMENT





LUDMILA GAVRILIUC





BIOCHEMISTRY
Lectures for students of Medical Departments















2


Chisinau
Editorial-Polygraphic Center Medicina
2011
CZU 577.1 (076.5)
G26

Aprobat de Consiliul metodic central cu nr.1 din 05.11.2010



Autor: Gavriliuc Ludmila profesor universitar catedrei Biochimie si Biochimie clinic,
doctor habilitat n medicin

Recenzeni: Vovc Victor profesor universitar, doctor habilitat in medicin,
sef catedrei Fiziologie omului
Cemortan Igor conferentiar universitar, doctor in biologie,
sef catedrei Biologie molecular si Genetica uman


Panciuc Liliana - lector universitar catedrei Limbi moderne
Costin Rodica lector superior universitar catedrei Limbi moderne






Redactor:

Corector:

Machetare computerizat:

















Medicina, 2011
DESCRIEREA CIP A CAMEREI NAIONALE A CRII
3





INTRODUCTION

Biochemistry can be defined as the science concerned with the chemical basis of life. The cell is
the structural unit of living systems. Thus, biochemistry can also be described as the science concer-
ned with the chemical constituents of living cells and with reactions and processes they undergo. By
this definition, biochemistry encompasses large areas of cell biology, of molecular biology, and of
molecular genetics.
The major objective of biochemistry is the complete understanding, at the molecular level, of all
of the chemical process associated with living cells. To achieve this objective, biochemists have sou-
ght to isolate the numerous molecules found in cells, determine their structures, and analyze how they
function. Many techniques have been used for these purposes.
Knowledge of biochemistry is essential to all life sciences. The biochemistry of the nucleic acids
lies at the heart of genetics; in turn, the use of genetic approaches has been critical for elucidating
many areas of biochemistry. Physiology, the study of body function, overlaps with biochemistry
almost completely. Immunology employs numerous biochemical techniques, and many immunologic
approaches have found wide use by biochemists. Pharmacology and pharmacy rest on a sound know-
ledge of biochemistry and physiology; in particular, most drugs are metabolized by enzyme-catalyzed
reactions. Poisons act on biochemical reactions or processes; this is the subject matter of toxicology.
Many workers in microbiology employ biochemical approaches almost exclusively.
Biochemical approaches are being used increasingly to study basic aspects of pathology (the stu-
dy of disease), such as inflammation, cell injury, and cancer. Biochemistry and medicine are intimate-
ly related. Health depends on a harmonious balance of biochemical reactions occurring in the body,
and disease reflects abnormalities in biomolecules, biochemical reactions, or biochemical processes.
These relationships are not surprising, because life as we know it depends on biochemical reactions
and processes. Biochemical approaches are often fundamental in illuminating the causes of diseases
and in designing appropriate therapies. The judicious use of various biochemical laboratory tests is an
integral component of diagnosis and monitoring of treatment.
This book can be very useful to the students of Medical Departments: General Medicine, Sto-
matology, and Pharmacy. Biochemistry integrates and summarizes the essentials of medical bioche-
mistry for students in the health-related professions.
Anything more than an extremely superficial comprehension of life in all its diverse maknifes-
tations demands the knowledge of biochemistry.
The major objective of Biochemistry is the complete in all its diverse manifestations demands
a knowledge of Biochemistry.
Medical students who acquire a sound knowledge of Biochemistry will be in a position to con-
front, in practice and research, the 2 central concerns of the health sciences:
1. the understanding and maintenance of health;
2. the understanding and effective treatment of disease.

4














LECTURE 1

Subject: INTRODUCTION IN BIOCHEMISTRY. AMINO ACIDS

Biochemistry or Biological chemistry is the branch of knowledge that deals with the structure of
chemical compounds that make up part of living matter, their transformations and physico-chemical
processes that constitute the basis of vital activity. Biochemistry is a part of Biology; it encompasses
the areas that require physico-chemical and chemical approaches, methods, and techniques. The speci-
ficity of Biochemistry is easily seen from its name, which suggests the chemical basis of this discipli-
ne as well as the important role played by functional (biological) studies of the chemical processes that
occur.
Historically, Biochemistry is intimately related to Organic Chemistry, which deals with the che-
mical properties of compounds that make part of living matter, and to Physiology, which deals with
the functions of living organisms. The terms physiological chemistry and biochemistry as equiva-
lent concepts were therefore not accidental.
Biochemistry owes its formation to numerous contiguous disciplines and remains closely related
to them in the study of animate nature. Nonetheless, it is an original and independent branch of know-
ledge whose major objectives are the investigation of the structural and functional interrelationships
and conversions of chemical compounds in the living organism, the routes of energy transformation in
the living systems, the regulatory mechanisms of chemical conversions and physico-chemical proces-
ses in cells, tissues, and organs, the molecular mechanisms of the transfer of genetic information in li-
ving organisms, etc.

Proteins. Introduction to protein chemistry
Proteins (from the Greek proteios, primary) are the major cell components of any living orga-
nism. Proteins play the most important role in all biological processes.
Proteins are distributed among subcellular structures not in a uniform manner: in highest amounts
they occur in the cell sap (hyaloplasm, cytosol). The protein level in organelles is rather determined by
the size and number of the organelles in a cell. Proteins are high-molecular nitrogen-containing orga-
nic compounds with complex structural organization, polymers composed of amino acids linked into
chains by covalent peptide bonds. Protein monomers are o-amino acids of the L-series (levorotatory).
High molecular mass is a very important characteristic of proteins. Depending on the chain length
all polypeptides are conventionally classified into peptides (containing from 2 to 10 amino acids),
polypeptides (from 10 to 40 amino acids), and proteins (over 40 amino acids). The molecular mass for
peptides is close to 1000, for polypeptides to 4000, and that for proteins from 4000-5000 to a few 10
6
.
For example:
glucagon 4 000
insulin 6 000
trypsin 23 800
glutamate dehydrogenase 1 000 000.

5
Amino acids as structural monomers for proteins
Amino acids, or aminocarboxylic acids, are organic carboxylic acids in which at least one hydro-
gen atom of the hydrocarbon chain is replaced by an amino group. L-Amino acids are classified into
o-, |-, -types, depending on the position of the carbon bearing an NH
2
group with respect to
COOH group. About 200 various amino acids have been identified in differrent species of the animate
nature. About 60 different amino acids and their derivatives are found in the human organism; still, not
all of them serve as constituents for proteins.
Amino acids are 20 which are as constituents of proteins. Amino acids are divided into two gro-
ups: proteogenic, or proteinogenic, amino acids, which are normally components of proteins, and
nonproteogenic (nonprotein) amino acids, usually not incorporated into proteins.
Among the proteogenic amino acids, there are major amino acids (in total number of 20) and rare
amino acids. Actually, the rare protein amino acids (for, example, hydroxyproline, hydroxylysine,
aminocitric acid, etc.) are derivatives of the 20 major amino acids.
Other amino acids do not participate in the protein synthesis; they occur in the cells either in a
free state (as metabolites), or are part of nonprotein compounds.
-Aminobutyric acid occurs in a free state and functions as an inhibitory neurotransmitter; |-ala-
nine is a component of a vitamin, pantothenic acid.
Structure and classifications of proteogenic amino acids
All protein amino acids are L-amino acids bearing an NH
2
group on the carbon at o-position:

o
R-CH-COOH

NH
2

Three classifications of amino acids are currently adopted:
1. Structural classification, based on the structural features of side-chain radicals of amino acids.
2. Electrochemical classification, based on the acid-basic properties of amino acids.
3. Biological classification, based on the functional priority of amino acids for the organism.
According to their electro-chemical (or acid-basic) properties, amino acids are divided into three
groups: acidic, basic and neutral, depending on the physico-chemical properties of the side-chain radical R.
Acidic amino acids are those having additional carboxylic groups in the side-chain radical, which
provide for enhanced acidic properties in this group of amino acids: aspartic, glu-tamic.
Basic amino acids include lysine, arginine, histidine, i.e. amino acids carrying an additional basic
group (amino) contributing to their enhanced bacic properties.
Neutral amino acids are the remaining acids. Their side-chain radicals exhibit neither acidic, nor
basic properties.
Depending on the side-chain radical polarity, amino acids of the two former groups (acidic and
basic) belong to polar; and amino acids of the third group (neutral) to nonpolar, or hydrophobic, acids.
According to their biological or physiological importance, amino acids are also subdivided into
three groups: essential, half-essential, and nonessential.
Essential amino acids cannot be synthesized in the organism from other compounds; therefore
they must be supplied to the organism as taken with the food. For the human organism, eight amino
acids are absolutely essential. These are acids: valine, leucine, isoleucine, threonine, lysine, methioni-
ne, phenylalanine, and tryptophan.
Half-essential amino acids are formed within the organism, but not in sufficient amounts; therefo-
re, they must partly be supplied in food. For the human organism, such amino acids are arginine, tyro-
sine, and histidine.
Nonessential amino acids are synthetized by the organism in adequate amounts from essential
amino acids or other compounds. The remaining amino acids are included among nonessential amino
acids. They are: glycine, alanine, serine, cysteine, cystine, aspartic acid, glutamic acid, and imino acids
proline, hydroxyproline.

Structural classification, based on the structural features of side-chain radicals

glycine aspartic acid threonine isoleucine
6


histidine lysine methionine tryptophan


serine proline phenylalanine

Figure 1. Structure of amino acids

The amino acids provide material for the synthesis of such important constituents as proteins
which are basically indipensable for the perpetuation of life.

Physical and chemical properties of amino acids
1. Acid-basic properties of amino acids
Viewed from the chemical standpoint, amino acids are amphoteric electrolytes, i.e. they exhibit
properties of both an acid and a base. The acidic groups of amino acids are: carboxylic group (-
COOHCOO
-
+ H
+
) and protonated o-amino group (-NH
3
+
NH
2
+ H
+
). Basic groups of amino
acids are: dissociated carboxyl group (-COO
-
+ H
+
COOH) and o-amino group (-NH
2
+ H
+

NH
3
+
).
Amino acids in aqueous solutions have been shown to occur as a dipolar species, or zwitterion.

R-CH-COOH Neutral form
|
NH
2

R-CH-COO
-
R-CH-COOH Dipolar form
| |
NH
2
NH
3
+


R-CH-COO
-
|
NH
3
+
Transition form

2. Effect of the medium pH on ionization of amino acids
The change of medium pH from acidic to basic affects the charge on dissolved amino acids. In
acidic medium (pH<7), all amino acids carry a positive charge (i.e. they occur as cations), since an
excess in environmental protons suppresses the dissociation of the carboxyl group:

R-CH-COO
-
+ H
+
R-CH-COO
-

| |
NH
3
+
NH
3
+

In acidic medium under applied electric field, amino acid molecules are carried over to the catho-
de (-). In basic medium (pH>7) with an excess of OH
-
ions, amino acids occur as negatively charged
species (anions), the NH
3
+
group being subject to dissociation:

R-CH-COO
-
+ OH
-
R-CH-COO
-
+ H
2
O
| |
NH
3
+
NH
2

In this case, under applied electric field amino acid molecules move towards the anode (+). The-
refore, amino acids, depending on the medium pH, can carry net zero, positive, or negative charges.
7
The state in which the net charge on an amino acid is equal to zero is referred to as isoelectric.
The value of pH at which such a state is attained, when the amino acid molecules are at rest under app-
lied electric field without preferential displacement neither to anode, nor to cathode, is named the iso-
electric point and is denoted pH. The isoelectric point is a very accurate indicator of acid-basic proper-
ties for various functional groups in an amino acid and is an important feature characteristic of the
amino acid. The isoelectric points for nonpolar (hydrophobic) amino acids are close to the neutral
value of pH (from 5,5 for phenylalanine to 6,3 for proline); for acidic amino acids, it has lower values
(e.g. 3,2 for glutamic acid and 2,8 for aspartic acid). The isoelectric point for the major amino acids,
histidine and, especially, lysine and arginine, is significantly greater than 7.
3. Stereoisomerism of amino acids
All proteogenic amino acids, excepting glycine, possess at least one asymmetric carbon atom
(C*) and exhibit optical activity, most of them being levorotatory. They exist as spatial isomers, or
stereoisomers. The stereoisomers are referred to the L- or D-series according to the arrangement of
substituents around the asymmetric carbon atom.
All the protein-constituting amino acids belong to the L-isomeric series.
Formerly it was believed that D-amino acids do not occur in the animate nature. D-amino acids
have been found in the bacteria and antibiotics generated by microorganisms.
In all the amino acids (except glycine) the carbon atom attached to nitrogen and alpha (o) to the
carboxyl group is asymmetric. It has been found that in general the configuration of the asymmetric
group of the naturally occurring amino acids is the same as the configuration of this group in L-glyceric
aldehyde to which the configuration of the L-series of sugars is referred.

CH=O COOH COOH
| | |
HO CH H
2
N-CH H
2
N-CH
| | |
CH
2
OH R CH
3

L-glyceric aldehyde L-amino acid L-alanine
Nomenclature for peptides and polypeptides
Peptide is defined as a linear peptide chain that is composed of amino acid residues linked
through peptide bonds. The high stability of the structure is provided by the covalent peptide bonds
that are formed by o-amino group of one amino acid and the o-carboxyl group of the neighbouring
amino acid:
O O
|| ||
H
2
N-CH-C-OH + H-N-CH
2
-COOH H
2
N-CH-C-N-CH
2
-COOH
| | -H
2
O | |
CH
3
H CH
3
H

alanine glycine alanylglycine (ala-gly)

Peptide linkage formed by condensation of the carboxyl of one amino acid and the amino group
of another with the elimination of water. The peptide bond formed with the involvement of the imino
group of proline (R) or hydroxyproline takes a different configuration:

H O H O
| || | ||
H
2
N-C-C-OH + HN-R H
2
N-C-C-N-R
| | -H
2
O | |
CH
3
COOH CH
3
COOH

alanine proline ala-pro
The peptide bond is a repeating unit of the polypeptide chain. Open chains contain a free o-amino
group at one end (N-end) of the chain and a free o-carboxyl group at the other end (C-end):

N-end H
2
N-CH-C-NH-CH-C-NH-CH-COOH C-end
| || | || |
R O R O R
8

Nomenclature for peptides and polypeptides
The name of a peptide is composed of the names of constituent amino acids. Two amino acids
give a dipeptide, three amino acids give a tripeptide, etc. In naming a polypeptide, all of the amino
acids are enumerated in consecutive order, always starting with the N-terminal amino acid and fini-
shing with the C-terminal amino acid. Each amino acid is given the ending-yl in place of the most
common ending-inethe C-terminal amino acid retains its name unchanged. For example, the tripepti-
de ala-ser-met is named alanyl-seryl-methionine.

LECTURE 2

Subject: PROTEINS: STRUCTURE AND LEVELS OF STRUCTURAL
ORGANIZATION OF PROTEINS

Four levels of structural organization of proteins are recognized: primary, secondary, tertiary and
quarternary. Each level has its proper specificity.

Primary structure of protein
The primary structure of protein is defined as a linear polypeptide chain that is composed of
amino acid residues linked through peptide bonds. The primary structure is the simplest level of struc-
tural organization of any protein molecule. The high stability of the structure is provided by the cova-
lent peptide bonds that are formed by the neighbouring amino acid:

O O
|| ||
H
2
N-CH-C-OH + H-N-CH
2
-COOH H
2
N-CH-C-N-CH
2
-COOH
| | -H
2
O | |
CH
3
H CH
3
H

alanine glycine alanylglycine (ala-gly)

The peptide bond formed with the involvement of the imino group of proline (R) or hydroxypro-
line takes a different configurations:

H O H O
| || | ||
H
2
N-C-C-OH + HN-R H
2
N-C-C-N-R
| | -H
2
O | |
CH
3
COOH CH
3
COOH

alanine proline ala-pro
Methods for determination of protein primary structure are: acidic, basic and enzymic hydroly-
sis, ion-exchange chromatography coupled to amino acid analyzer, sequencing (as performed on sequ-
encers).

Secondary structure of protein
The secondary structure refers to the way the peptide is folded into an ordered structure owing to
hydrogen bonding between peptide groups of the same chain or juxtaposed polypeptide chains. By
configuration the secondary structures are classified into helical structures (o-helix) and pleated sheets
(|-structure and cross-|-form).
o-Helix. This type of secondary structure, which resembles a regular helix, is formed owing to
hydrogen bonds between peptide groups within the same polypeptide chain. The o-helical structural
model, which takes into account all known properties of the peptide bonds, was proposed by Pauling
and Cori.





9








Figure 2. Secondary structure of protein (o-structure)

The main features of the o-helix are:
1. Helical configuration of the polypeptide chain of screw-type (rotation-translational) symmetry.
2. Formation of hydrogen bonds between the peptide groups of every first and fourth amino acid
residues.
3. Regularity of the turns along the helix length.
4. Equivalence of all amino acid residues in the o-helix, irrespective of the structure of their side-
chain radicals.
5. Nonparticipation of the amino acid side-chain radicals in hydrogen bonding, i.e. in the forma-
tion of o-helix.
The o-helix resembles a slightly distended heating coil. The regularity of hydrogen bonds bet-
ween every first and fourth peptide groups determines the regularity of the polypeptide chain turns.
The axial pitch of o-helix is 0.54 nm; it contains 3.6 amino acid residues.
|-Structure. This variety of the polypeptide chain secondary structure adopts a slightly bent con-
figuration and is formed by means of interpeptide hydrogen bonds within the same polypeptide chain
or with involvement of hydrogen bonds between juxtaposed polypeptide chains. |-structure is also
referred to as a pleated sheet structure. Several types of |-structure are known. Finite pleated lengths
of a single peptide chain are called cross-|-forms (short |-structures). In the cross-|-form, hydrogen
bonds are formed between the peptide groups of polypeptide chain loops. Another type of |-structure
is characteristic of the whole polypeptide chain, which is held is a stretched state by hydrogen bonds
between juxtaposed parallel polypeptide chains.
In shape, this structure resembles the bellows of an accordion. |-Structures can be formed by pa-
rallel chains (with N-termini of the polypeptide chains pointing in the same direction), or by antiparal-
lel chains (with the N-termini pointing in the opposite direction). The side-chain radicals of one sheet
are located between the side-chain radicals of the other sheet.


Figure 3. Secondary structure of protein (|-structure)

Owing to the reorganizable hydrogen bonds in proteins the o-structure can be reversibly conver-
ted to the |-structure. The regular interpeptide hydrogen bonds by means of which a polypeptide chain
retains its o-helical conformation become replaced by interchain hydrogen bonds between the stret-
ched fragments of neighbouring uncoiled polypeptide chains. Such a transition has been established in
the hair protein, keratin.
Other types of bonds contribute, but little to the secondary structure formation, with the exception
of disulphide bonds formed at the sites along the polypeptide chain where cysteine residues are pre-
sent. Short peptides, owing to the disulphide bonds, become closed to rings.
Numerous proteins possess both o-helical regions and |-structures. Proteins exhibit a varied ex-
tent of helicity. A high percentage of o-helical structures is observed in paramyosin, myoglobin, and
hemoglobin. In contrast, a sizeable portion of the polypeptide chain in trypsin and ribonuclease is
10
pleated in sheet |-structures. The proteins of supporting tissues, viz. keratin (hair and wool protein),
collagen (skin and tendon protein) and fibroin (natural silk protein) all possess a |-configuration of
their polypeptide chains.
Methods for determination of protein secondary structureare: spectropolarimetry (measurement
of rotation angle for linearly polarized light on spectrophotometers), isotope exchange method, UV
spectrophotometry (measurement of protein UV absorption at 190 nm on spectrophotometers), IR spec-
troscopy (measurement of infrared absorption spectra of proteins using infrared spectrophotometers).

Tertiary structure of proteins
The tertiary structure of protein is referred to as a specific mode of spatial arrangement of the
polypeptide chain. In regard to their tertiary structure, the proteins are chiefly divided into globular
and fibrous (or fibrillar) species. Globular proteins have most commonly an ellipsoid shape, while fib-
rous proteins are elongated (rodor spindle-like).
Bonds contributing to stabilization of tertiary protein structure. Bonds that are formed between
the side-chain radicals of amino acids play a role in the stabilization of the tertiary structure. These
bonds may be classified into strong (covalent) and weak (polar, van-der-Waals) bonds.
Covalent bonds comprise disulphide bonds (-S-S-) between the side chains of cysteine residues












Figure 4. Tertiary structure of protein (B, myoglobin)

located in different parts of polypeptide chains; isopeptide (or pseudopeptide) bonds between the ami-
no groups of the side chains of lysine or arginine and the COOH side-chain groups of aspartic or glu-
tamic acids. This explains the name (peptide-like) conferred on the bonds of this kind. Of rare occur-
rence is the ester bond formed with the involvement of a COOH group of dicarboxylic amino acids
(aspartic and glutamic) and the OH group of hydroxyamino acids (serine and threonine).
Polar bonds comprise hydrogen and ionic bonds. Hydrogen bonds commonly arise between
NH
2
, -OH, or SH side-chain groups of one amino acid and carboxylic group of the other amino acid.
Ionic, or electrostatic, bonds are formed between charged side-chain groups NH
3
+
(in lysine, arginine,
histidine and COO
-
(in aspartic and glutamic acids) brought into a closer contact.
Nonpolar, or van-der-Waals, bonds are formed between hydrocarbon radicals of amino acids.
The hydrophobic radicals of alanine, valine, isoleucine, methionine, and phenylalanine interact in
aqueous medium. Weak van-der-waals forces favour the formation of a hy-drophobic core composed
of nonpolar radicals in the interior of a protein globule. The contribution of van-der-waals forces to the
architecture of polypeptide chains increases with the number of nonpolar amino acids.
The multiplicity of bonds between the side-chain radicals of constituent amino acids determines
the spatial configuration of a protein molecule. The conformation of the polypeptide chain tertiary
structure is determined by the properties of the side-chain radicals of constituent amino acids (which
exert no sizeable influence on the formation of primary and secondary structures) and those of the
microenvironment, i.e. medium. In folding, the protein polypeptide chain tends to adopt an energeti-
cally favorable configuration corresponding to a minimum of free energy.
In protein molecules with tertiary structure, there occur regions formed as o-helices, |-structures
(pleated sheets) and random coils. It is only a proper spatial arrangement that renders the protein acti-
ve; otherwise a disorganization of its spatial structure produces alterations is its properties and may
lead to a loss of biological activity.
Methods for determination of tertiary protein structure are: electron microscopy, X-ray structu-
ral analysis.

11
Quarternary structure of proteins
Proteins composed of a single polypeptide chain possess only a tertiary structure. They include
myoglobin, a muscle tissue protein involved in oxygen binding, as well as a number of enzymes (liso-
zyme, pepsin, trypsin, etc.). However, certain proteins are built of several polypeptide chains, each
chain of which has a tertiary structure.
For such proteins, the notion of a quarternary structure has been suggested. The quarternary
structure presents itself as an aggregation of two or more polypeptide chains with a tertiary structure
organized into a single functional protein molecule. A protein with a quarternary structure is referred
to as an oligomer and its polypeptide chain with a tertiary structure are referred to as protomers, or
subunits.
At the quarternary level of organization, proteins retain the main configuration of their tertiary
structure (globular or fibrous). For example, hemoglobin, a protein exhibiting a quarternary structure,
is composed of four subunits. Each of the subunits is a globular protein, and hemoglobin also has an
overall globular configuration.




Quarternary structure of hemoglobin (Hb: 2o and 2| subunits)

Stabilization of protein quarternary structure
All proteins exhibiting a quarternary structure are amenable to isolation as individual macromole-
cules resisting dissociation into the constituent subunits. Numerous ionic, hydrogen and, occasionally,
disulphide bonds are formed between the subunit polar groups providing thereby for the persistence of
the involved subunits as an organized tightly bound complex.
Methods for determination of protein I V structure are: electron microscopy, X-ray structural
analysis , and electrophoresis.
Specific features of structural organization in certain fibrous proteins. The structural organiza-
tion of fibrous proteins reveals a number of specific features as compared with that of globular pro-
teins. These features may be exemplified by keratin, fibroin, and collagen. Keratins exist in - and -
conformations. o-Keratin and fibroin both possess a secondary pleated sheet structure; still, in keratin,
the polypeptide chains are parallel and in fibroin, antiparallel. The fibrous protein is typified by colla-
gen, a major protein in the human organism (about
1
/
3
of total protein mass). Collagen is responsible
for the strength and low flexibility of connective tissue and is found in bones, tendons, skin, teeth, etc.
In collagen, glycine accounts for one third, and proline or hydroxyproline, for about one fourth of the
total of amino acid residues. Polypeptide chain of collagen resembles a broken line. It contains about
1000 amino acids and has a molecular mass of the order of 10,000. The polypeptide chain is built of
repeating units, each unit being composed of tree amino acids (triplet) glycine-proline-hydroxyproli-
ne. Proline, hydroxyproline, and glycine (antihelical amino acids) hinder the formation of -helices.
Therefore, three chains form a kind of twisted helices similar to three threads wound around a cylin-
der. The three helical -chains form a repeating collagen structure referred to as tropocollagen. Tropo-
collagen, by its organization, is a tertiary structure for collagen and a subunit for collagen fibrils. The
folding of tropocollagen subunits into a collagenic quaternary structure is accomplished stepwise. Sta-
bilization of collagenic structures is effected through the agency of interchain ionic, hydrogen, and
van-der-Waals bonds, with a minor participation of covalent bonds.

12
LECTURE 3

Subject: PHYSICAL AND CHEMICAL PROPERTIES OF PROTEINS

The physico-chemical properties of a protein are determined by its amino acid composition and
spatial organization. Proteins exhibit acid-basic, buffering, colloidal, and osmotic properties.

Proteins as amphoteric macromolecules
Proteins are amphoteric polyelectrolytes, i.e. they combine, similar to amino acids, acidic and ba-
sic properties. However, the nature of the constituent groups imparting amphoteric properties to pro-
teins differs to a significant extent from that of amino acids. The acid-basic properties of amino acids
are primary due to occurrence of o-amino and o-carboxyl groups (i.e. acid-base pairs) in them. The
amphoterism of proteins is due to the acid-base groups of side-chain radicals of protein-constituting
amino acids. It stands to reason that each molecule (polypeptide chain) of a native protein possesses a
minimum of one terminal o-amino and one terminal o-carboxyl group (providing the protein displays
a tertiary structure only). For a protein with a quaternary structure, the number of terminal -NH
2
and
COOH groups is equal to the number of subunits, or protomers. However, the small number of these
groups does not suffice to explain the amphoterism of protein macromolecules. Since the majority of
the polar groups are located on the surface of globular proteins, precisely these groups provide for the
acid-basic properties and the charge of a protein molecule. Acidic amino acids (aspartic, glutamic) are
responsible for acidic, and basic amino acids (lysine, arginine, and histidine), for basic properties of
proteins. Acidic properties of a protein become more pronounced, as the number of its constituent aci-
dic amino acids increases; accordingly, basic properties manifest themselves to a greater extent with
an increased number of basic amino acids. Poorly dissociating SH-groups of cysteine and the phenolic
group of tyrosine (formally treated as weak acids) practically do not affect the amphoterism of pro-
teins.

Buffering properties
Proteins, while exhibiting buffering properties, do not, nevertheless, possess any high buffering
capacity at physiological values of pH. An exception to the rule are proteins containing a large number
of histidine residues, since it is only the side chain of histidine that exhibits buffering properties within
a pH range close to the physiological pH. Such proteins are quite scarce, though. Hemoglobin is actua-
lly the only protein with a histidine content reaching 8% and is, therefore, a powerful intracellular buf-
fer in the erythrocytes which is capable of maintaining the blood pH value at a constant level.
The charge on a protein molecule is dependent on the contents of acidic and basic amino groups
in the side-chain radicals of these amino acids. The dissociation of COOH groups of acidic amino
acids causes a negative charge to develop on the protein surface, while the side-chain radicals of basic
amino acids carry a positive charge (owing to the addition oh H
+
ions to the basic groups). In a native
protein molecule the charge distribution is asymmetric, depending on the way the polypeptide chain is
folded in space. Acidic amino acids being in preponderance over basic amino acids in a protein, the
protein molecules acquire a net negative charge, i.e. become polyanions, and vice versa, if basic amino
acids prevail, the protein molecules develop a positive charge and behave as polycations.
Undoubtedly, the net charge on the protein molecule is dependent upon the pH of the medium: in
an acidic medium the charge is positive, and in a basic medium, negative. The value of the pH at
which a protein exhibits a net zero charge is referred to as the isoelectric point for the given protein. At
this point the protein is incapable of migrating under an applied electric field. The knowledge of the
isoelectric point is of almost importance for understanding the stability of proteins in solution, since in
the isoelectric state, proteins are the least stable. Uncharged protein particles may coalesce and preci-
pitate from solution.

Colloidal and osmotic properties of proteins
The behavior of proteins in solution displays a number of specific features. Common colloidal
solutions are stable only in the presence of a stabilizer whose molecules are adsorbed at the solute-
solvent interface, preventing thereby the colloid coagulation.
Aqueous solutions of proteins are stable and equilibrated; they do not precipitate with time (do
not coagulate) and do not require the presence of a stabilizer. Protein solutions are homogeneous and
may be referred to as true solutions. However, the high molecular mass of proteins imparts a number
of properties typical of colloidal systems to their solutions:
13
- characteristic optical properties (opalescence of solutions and ability to scatter the visible
light);
- low diffusion rate;
- inability to pass across semipermeable membranes;
- high viscosity;
- propensity to gelation.

Optical properties of proteins
Protein solutions, especially concentrated ones, exhibit a characteristic opalescence. With a pro-
tein solution irradiated sideways, the light beam becomes visible as a luminescent cone; this phenome-
non is known as the Tyndal effect. This light-scattering effect is explained by light diffraction due to
protein particles suspended in solution. It is commonly recognized that protein in the cellular proto-
plasm occurs as a colloidal solution, or sol. The ability of proteins and other biological molecules
(nucleic acids, polysaccharides, etc.) to scatter the light is made use of in the microscopic studies of
cellular structures: in the visible microscope, colloidal particles are apparent against the dark back-
ground as light spots in the cytoplasm.
The light-scattering properties of proteins and other high-molecular compounds are also used in
the quantitative determination by the nephelometric method: the light intensities scattered by suspen-
ded particles in the sample and reference solutions are compared.
Low diffusion rate. Proteins are characterized by limited diffusion rates as compared with com-
mon molecules and ions capable of moving at the rates higher over those for proteins by a few orders
of magnitude. The diffusion rate for proteins is influenced by the molecular shape of protein particles
rather than by their molecular mass. Globular proteins in aqueous solutions are more mobile than fib-
rous proteins. The intracellular distribution of proteins is effected via diffusion. Since protein diffusion
proceeds at a slow rate, it happens to be a rate-limiting factor for other processes dependent on the bio-
logical function of diffusing protein in respective regions of the cell.

Osmotic properties of proteins
Proteins, owing to their high molecular mass, are incapable of diffusing across semipermeable
membranes, whereas low-molecular compounds pass easily through such membranes. This property of
proteins is made use of in practice for purifying protein solutions from low-molecular contaminants.
This process is referred to as dialysis.
Biological membranes are likewise impermeable to proteins, therefore the osmotic pressure pro-
duced by protein is dependent both on the protein concentration inside and outside the cell. The osmo-
tic pressure due to protein is also referred to as the oncotic (produced by swellling) pressure.

High viscosity of protein solutions
High viscosity is characteristic not only of protein solutions, but also of high-molecular com-
pound solutions in general. As the protein concentration in solution becomes greater, the viscosity of
the solution increases due to a concomitant increase in colligative forces between the protein molecu-
les. The viscosity is dependent on the shape of molecules. Solutions of fibrous proteins are always
more viscous than globular protein solutions. Temperature and the presence of electrolytes are the fac-
tors that strongly affect the protein solution viscosity. As the temperature rises, the protein solution
viscosity decreases. The viscosity of a protein solution may increase to such an extent that the solution
loses fluidity and converts to a gel-like state.

Aptitude of proteins for gelation
Interaction of protein macromolecules in solution may lead to the formation of structural net-
works with water molecules trapped inside them. Such structurized systems are referred to as gels or
jellies. It is recognized that the protein of cell protoplasm can be converted to a gelatinous state. A de-
monstrative example is the body of a medusa which may be regarded as a living jelly with water con-
tent in it up to 90%.

Hydration of proteins and factors affecting protein solubility
Proteins are hydrophilic materials. A dry protein, when submerged in water, starts swellling
immediately as any high-molecular hydrophilic compound; with time, the protein molecules gradually
start passing into solution. In the course of swelling, water molecules penetrate into the interior of pro-
tein structure of polypeptide chains becomes loose. Further water intake leads to the detachment of
14
individual protein molecules from the protein bulk and to their passage into a state of dissolution. Still,
the dissolution is not necessary sequent upon swellling; certain proteins, for example, collagen, persist
in the swollen state even after having taken up, a large amount of water.

Factors affecting protein solubility
Solubility of proteins varies over a wide range. The solubility is determined by amino acid com-
position (polar amino acids impart a greater solubility to proteins over nonpolar amino acids), specifi-
city of structural organization (globular proteins are, as a rule, more soluble than fibrous proteins), and
the properties of a solvent.
The charge on protein molecules and the hydration shell around them impart stability to the
protein solutions. Each macromolecule of an individual protein carries a net charge of the same sign,
which prevents agglutinations of the protein molecules in solution and their precipitation. Any factor
contributing to the conservation of charge and hydration shell favors the solubility of proteins and
their stability in solution. There is a close relationship between the charge on a protein (or the number
of polar amino acids in it) and its hydration properties: the higher the number of polar amino acids in
the protein, the greater the amount of water it is capable of binding (per 1g of dry weight). A protein
contains a large number of cationic and anionic groups; nonetheless, its solubility in water is rather
poor. The intermolecular salt bridges formed make protein particles coagulate and precipitate from
solution.
What environmental factors affect the solubility of proteins and their stability in solution?
Neutral salts, acids, alkalis, organic solvents (ethanol, acetone), alkaloids, heavy metal salts (mer-
cury, copper, zinc, etc.). The process of protein precipitation by a neutral salt solution is called salting-
out. A specific feature in the behaviour of proteins obtained by salting-out is the retention of their
native biological properties after the salt has been removed. The salting-out mechanism is operable in
that the anions and cations of the added solution destroy the protein stability. It is probable also that si-
multaneously neutralization of the protein charge by salt ions takes place, which facilitates precipitati-
on. The salting-out is widely used for separation and purification of proteins.


Chemical Precipitation

Denaturation (denativation) and renaturation (renativation) of proteins
Certain agents destroy the higher levels of structural organization of protein molecules (seconda-
ry, tertiary, and quarternary) with the retention, however, of the primary structure, and the protein thus
loses its native physico-chemical, and, what is important, biological property. This phenomenon is
referred to as denaturation (denativation). It is characteristic of the protein molecules that possess a
complex spatial organization. Synthetic and natural peptides are incapable of denaturation.
Denaturation is accomplished through breakdown of the bonds that stabilize the quarternary, ter-
tiary, and occasionally, secondary structures. The polipeptide chain unfolds and remains in solution in
the unfolded state, or as a random coil. Macromolecules become stripped of the hydration shells, and
the protein separates from solution as a precipitate. However the precipitated denatured protein differs
markedly from the precipitated form of the same protein produced by salting-out, since in the former
case native properties of the protein are lost and in the latter, retained. This implies that the mecha-
nisms operable in denaturation and salting-out are different. The salting-out leaves the protein native
structure unaffected, whereas the denaturation destroys it.
15
The factors producing denaturation are differentiated into physical and chemical ones. Physical
factors are: temperature, pressure, mechanical action, and ultrasonic and ionizing irradiation. Heat de-
naturation of proteins has been studied more thoroughly than other processes and has been recognized
as a characteristic feature of proteins. It was since long known that proteins, when heated, curdle (coa-
gulate) to form a precipitate. Most proteins are thermolabile, or heat-modifiable; still, there are pro-
teins known to be very resistant to heat. For example, such are: trypsin, lysozyme, chymotripsin and
certain biomembrane proteins.
Chemical factors eliciting denaturation are acids and alkalis, organic solvents (ethanol, acetone),
detergents (cleansing agents), certain amides (urea), alkaloids, and heavy metal salts (mercury, copper,
zinc, etc.). The mechanism of denaturation by chemical agents is dependent on their physico-chemical
properties. Acids and alkalis are widely used as protein precipitants, or coagulators. In the stomach of
humans and animals, the natural denaturating agent, hydrochloric acid, is secreted, which denatures
proteins and facilitates their enzymic degradation.
Denaturation was for a long time believed to be an irreversible process. In certain occasions, ho-
wever, the denatured protein is capable of restoring its biological activity after the denaturation agent
has been removed. The process of recovering by the protein of its physico-chemical and biological
properties is called renaturation, or renativation. If the denaturated protein (after removal of denatu-
rants) is again self-organized to its initial structure, its activity also becomes restored.
Methods for purification of proteins are: mechanical grinding (homogenization), differential
centrifugation, treatment with agents disjoining intercellular contacts, extraction, salting-out, acid-base
treatment, dialysis, ultrafiltration; ion-exchange, adsorption, gel, affinity chromatography; electropho-
resis, isoelectrical focusing in pH gradient, etc.

LECTURE 4

Subject: ENZYMES: CLASSIFICATION, NOMENCLATURE AND
CHEMICAL STRUCTURE OF ENZYMES, COENZYMES

Enzymes are biological catalysts of protein nature. The term enzyme (from the Greek en, inzy-
me, leaven) was first coined in the early 17
th
century by the Holland physician van Helmont for sub-
stances affecting fermentation. The enzyme is a synonym of the term ferment (from the Latin fer-
mentum, leaven). The enzymes are peculiar to the animate nature only and are used as specific regula-
tors of metabolism. No chemical process in the living organisms can be effected without involvement
of enzymes.
At the present time, the enzymology is an extremely important, rapidly growing branch of bioche-
mistry, and its achievements are widely used in practical medicine, pharmaceutics, food industry, and
other related industries.

Common and distinct features in enzymes and nonenzymic catalysts
Enzymes and nonbiological catalysts, in obeying the general laws of catalysis, share the following
common features:
1. They catalyze energetically feasible reactions only.
2. They never alter the reaction route.
3. They go not affect the equilibrium of a reversible reaction, but rather accelerate its onset.
4. They are never consumed during the reaction. Therefore, a cellular enzyme functions until it
becomes impaired for one or another reason.
However, enzymes exhibit a number of features that distinguish them from nonbiological cata-
lysts. These distinctions are due to structural specificities of the enzymes which are complex protein
molecules.
1. The rate of enzymic catalysis is much superior to that of nonenzymic catalysis. It follows there
from that enzymes lower the activation energy of reactions to a greater extent as compared with non-
biological catalysts.
2. Enzymes exhibit a high specificity. There ae enzymes that act selectively on only one stereo
isomer of a compound. The high specificity of enzymes enables them to direct metabolic processes to
strictly defined channels.
3. Enzymes catalyze chemical reactions under mild conditions, i.e. at normal pressure, low tem-
perature (about +37
o
C), and pH close to that of the neutral medium (pH=71). This behaviour diffe-
rentiates them from other catalysts active at high pressure, extreme pH values, and high temperature.
16
Enzymes, because of their proteinic nature, are susceptible to temperature variations (i.e. are ther-
molabile) and to the change of medium pH.
4. Enzymes are catalysts with controllable activity, the behaviour never encountered in nonbiolo-
gical catalysts. This unique property in enzymes allows changing the rate of metabolism in the orga-
nism depending on the environmental conditions, i.e. adapting the metabolic activity to the action of
various factors.
5. The rate of an enzymic reaction is proportional to the amount of enzyme, while no strictly defi-
ned relationship of this kind is found in nonbiological catalysts. Therefore, the short supply of an en-
zyme in the living organism signifies a lower rate of metabolism and, on the contrary, the additional
production of an enzyme is one of the adaptive routes for the organism cells.
The highest energy position (peak position) represents the transition state. With the catalyst, the
energy required to enter transition state decreases, thereby decreasing the energy required to initiate
the reaction.

The relationship between activation energy (E
a
) and enthalpy of formation
(H) with and without a catalyst

Classification and nomenclature of enzymes
At present, it is believed that the cell contains about 10
4
enzyme molecules capable of catalyzing
over 2000 various reactions. There are 1800 enzymes known to date. About 150 enzymes have been
isolated in the crystalline form.
There are 2 classifications of enzymes:
1. The trivial (working) classification
The trivial name for an enzyme was made up of the ending ase added to the name of the substra-
te subject to the action of the enzyme in question: for example, saccharose + ase =saccharase
2. The systematic classification
The systematic name for an enzyme is constructed in a more complicated manner. It is made up of
the names of substrates of the chemical reaction catalyzed by the enzyme, the name of the type of the
catalyzed chemical reactions, and the ending ase. For example, the systematic name for the enzyme
lactate dehydrogenase is written as:

L-lactate: NAD
+
-Oxidoreductase
substrate 1 substrate 2 type of chemical reaction

The systematic names are given to the explore enzymes only. All the enzymes are classified into
6 groups; of these, each is assigned a definite number:
1. oxidoreductases
2. transferases
3. hydrolases
4. lyases
5. isomerases
6. ligases (or synthetases).
The name of the group indicates the type of the chemical reaction catalyzed by enzymes. Therefo-
re, there are six major types of enzymic reactions. The groups are divided into subgroups; the latter are
further subdivided into subsubgroups. According to the numerical classification system, each enzyme
receives a four-part number whose numerals are separated by a dot:
LACTATE DEHYDROGENASE E.C. 1. 1. 1. 27
Enzymatic classification ________________|
17
group number __________________________ |
subgroup number ____________________________|
subsubgroup number ____________________________|
ordinal number in subsubgroup _______________________|
All new enzymes are classified only in accordance with the recommendations of the Committee
on Enzyme Nomenclature of the International Union of Biochemistry.
Oxidoreductases are enzymes that catalyze redox reactions. The substrate subject to oxidation
with oxidoreductases is regarded as a hydrogen donor. For this reason, the enzymes in this group are
called dehydrogenases, or, less commonly, reductases. They play a decisive role in the energy metabo-
lism.
Transferases are enzymes that catalyze reactions of transfer of various moieties from one sub-
strate (donor) to another (acceptor). The enzymes that catalyze the transfer of methyl groups are called
methyl transferases; those that catalyze the amino group transfer are called amino transferases, etc.
Hydrolases are enzymes that catalyze the substrate bond cleavage by adding water. The trivial
names for hydrolases are made up by adding the ending ase to the name of substrate. Systematic na-
mes must, by convertion, contain the term hydrolase (for example, amylase, saccharase).
Lyases are enzymes that catalyze bond-cleaving reactions in a substrate without oxidation or ad-
dition of water (for example, pyruvate decarboxylase).
Isomerases are enzymes that catalyze structural rearrangements within a single molecule (for
example, triose isomerase).
Ligases (synthetases) are enzymes that catalyze the addition of two molecules using the energy
of phosphate bond. ATP or other nucleoside phosphates serve as energy sources in the synthetase-
catalyzed reactions (for example, DNA-ligase).
Structural and functional ogranization of enzymes
The enzymes exhibit all features characteristic of the protein structural organization. They possess
four levels of organization: primary, secondary, tertiary, and quaternary. The enzymes with quaternary
structure are composed of protomers (subunits) and constitute a preponderant type. Similarly to other
functional proteins, they are classified into simple enzyme proteins and conjugated (or compound)
enzyme proteins. A conjugated enzyme is composed of a protein portion, apoenzyme, and a nonprotein
portion, cofactor. The cofactors in enzymes are metal ions and coenzymes. Taken singly, the apoenzy-
me and cofactor exhibit a low (if any) activity as catalysts, but united, they make up a molecule of the
active enzymes, or holoenzyme.
Functional organization of enzyme
In the three-dimensional structure of a simple, as well as a conjugated, enzyme, there are distin-
guished a number of regions responsible for certain specific functions. A portion of the enzyme mole-
cule constitutes the active center (centre), i.e. the site in the enzyme spatial structure where the bin-
ding with a substrate (S) takes place (substrate is a compound that undergoes conversion by the action
of enzyme).
Alongside of the active center, an enzyme has a regulatory, or allosteric, center spatially remote
from the active center in the enzyme molecule. The name allosteric center (from the Greek allos,
other, or foreign) implies that the molecules bound to this center are structurally (sterically) dissimilar
from the substrate but exert influence on the binding and conversion of substrate at the active center
by changing the substrate configuration.
The enzyme molecule can have more than one allosteric center. Compounds capable of binding to
an allosteric center are called allosteric effectors. They exert influence, through the allosteric center,
on the function of the active center in a facilitating or an inhibitory manner. Accordingly, allosteric
effectors are referred to as positive (activators) or negative (inhibitors).
Structure of the active center
There are distinguished, in the active center, a contact site (or anchor site) for binding a substrate,
and a catalytic site at which the conversion of the bound substrate takes place. Usually, the enzyme
active center is made up of 12 to 16 amino acid residues of a polypeptide chain; occasionally, their
number may be larger.
Functional groups of enzyme active center
In simple enzymes, the role of functional groups at the contact and catalytic sites is assigned to
the side-chain radicals of amino acids only. In conjugated enzymes, the leading part in these processes
is played by cofactors.
18
The following functional enzyme groups take part in catalysis: -COOH, -NH
2
, -OH, -SH and
other.
Cofactors and their role in enzyme function
Cofactors are either bound to the enzyme active center, or susceptible to easy cleavage by dialy-
sis. The term prosthetic group is applied to tightly-bound cofactors, similar to the accepted definition
of the nonprotein portion in nonenzymic protein. Still, such a definition appears to be somewhat arbi-
trary, since the cofactor (usually coenzyme) can be tightly bound to the active center of one enzyme
and none, to that of another enzyme. Vitamins are parent materials for coenzymes, therefore, their die-
tary deficiency affects the synthesis of these coenzymes and leads, as a consequence, to an impaired
function of the corresponding conjugated enzymes.
Thiamine coenzyme is derived from thiamine (vitamin B
1
).



Figure 5. Structure of TPP (or TDP)



Figure 6. Nicotinamide coenzymes (structures of NAD and NADP)

Niacin (vitamin B
5
, nicotinic acid) serves as a source for generating nicotinamide coenzymes. The
latter species include nicotinamide adenine dinucleotide (NAD) and nicotinamide adenine dinucleotide
phosphate (NADP).
One of the mononucleotides of these coenzymes contains nicotinamide, the other one is represen-
ted by adenilic acid.
Pyridoxine coenzyme. Pyridoxine coenzyme is derived from pyridoxine (vitamin B
6
).

Figure 7. Coenzyme of vitamin B
6
, pyridoxal phosphate

Flavin coenzymes. These derived from riboflavin (vitamin B
2
) structurally related to the isoal-
loxazine derivatives. Coenzymes flavin mononucleotide (FMN) and flavin adenine dinucleotide
(FAD) are synthesized from riboflavin.
19



Figure 8. Chemical structures of vitamin B
2
and its coenzymes, FMN and FAD

Metalloporphyrin coenzymes include the hemes that participate as coenzymes in redox-reactions
catalyzed by oxidoreductases (cytochromes, catalase, peroxidase, etc.).
Metal ions are likewise capable of acting as cofactors. Metalloenzymes constitute a very wides-
pread group of enzymes accounting for 25% of all the enzymes. The role of the metal in these enzy-
mes may be quite different. Metalloenzymes are divided into two groups:
1. Enzymes in which the metal ion acts as an activator (these enzymes exhibit catalytic activity
in the absence of metal as well).
2. Enzymes in which the metal ion acts as a cofactor (in the absence of metal these enzymes are
inactive).

LECTURE 5

Subject: MECHANISM OF ENZYMIC ACTION. REGULATION OF
ENZYME ACTIVITY

The process of enzyme catalysis may conventionally be differentiated into three steps, each exhi-
biting its specificity.
1. Diffusion of a substrate to an enzyme resulting in a stereospecific binding of the former to the
active site of enzyme (formation of an enzyme-substrate complex ES).
2. Conversion of the primary enzyme-substrate complex into one or more activated enzyme-sub-
strate complexes (denoted ES* in the scheme).
3. Detachment of the reaction products from the active center of enzyme and their diffusion into
the environment (complex EP dissociates into E and P).

E + S ES ES* EP E + P



Diagrams to show the induced fit hypothesis of enzyme action

Specificity of enzyme action
20
Enzymes exhibit a varying degree of specificity towards substrates. Viewed from this standpoint,
the enzymes are subdivided into the following major types which are discussed in the order of their
decreasing specificity.
1. Absolute substrate specificity: the enzyme catalyzes conversion of a single substrate only. For
example, urease is active only in the conversion of urea.
2. Absolute group substrate specificity: the enzyme catalyzes conversion of a related group of
substrates. For example, alcohol dehydrogenase catalyzes conversion not only of ethanol, but other
alcohols too, though at different rates.
3. Relative substrate specificity: the enzyme catalyzes conversion of substrates belonging to
different groups of chemical compounds. For example, the enzyme cytochrome P
450
(hydroxylase) par-
ticipates in the hydroxylation of a large number (about 700) of compounds. Enzymes with relative
substrate specificity constitute the least specific enzymic system involved in the conversion of natural-
ly occurring materials, drugs, and toxins.
4. Relative group substrate specificity: the enzyme exhibits a specific activity towards individual
bonds within a group of substrates. For example, digestive enzymes (pepsin, trypsin) are specific to-
wards the peptide bonds formed between certain amino acid residues in various proteins.
5. Stereochemical substrate specificity: the enzyme catalyzes conversion of only one among all
possible substrate stereoisomers. This is an extreme case of enzymic specificity. For example, D-ala-
nine oxidase catalyzes the conversion of only D-alanine amino acid, and never of its stereoisomer, L-
alanine amino acid.
Kinetics of enzymic reactions
The kinetics of enzymic activity is a branch of enzymology concerned with the studies of enzy-
me-catalyzed reaction rates as affected by the chemical nature of substrate and enzyme, by the condi-
tions for their interaction, as well as by environmental factors. Otherwise stated, the enzyme kinetics
allows one to gain insight into the molecular mechanistic nature of the factors affecting enzymic cata-
lysis rates.
The rate of an enzymic reaction is defined by the amount of a material (or materials) converted
per unit time. The rates of such reactions are dependent on the environmental factors (temperature, pH
of medium, influence of native or foreign materials, etc.).
The principles of enzymic reaction kinetics have been laid down in the works of Michaelis and
Menten. The MichaelisMenten equation relates the initial reaction rate v
0
to the substrate concentra-
tion [S]. The corresponding graph is a rectangular hyperbolic function; the maximum rate is described
as v
max
(asymptote).


The MichaelisMenten equation describes the rates of irreversible reactions. A steady state solu-
tion for a chemical equilibrium modeled with MichaelisMenten kinetics can be obtained with the
GoldbeterKoshland equation.

Figure 9. Dependence activity of enzyme (V) on concentration of substrate (S)

The rate of an enzymic reaction is seen as a measure for catalytic activity of the enzyme involved
and is referred to simply as the activity of enzyme. The enzymic activity can be estimated only in an
indirect fashion, by a decrease in the amount of substrate converted, or by an increase in the concen-
21
tration of products formed per unit time. The dependence of reaction rate on the enzyme amount is li-
near in character.
Dependence of reaction rate on pH of medium
Usually, the curve relating the enzymic reaction rate to the medium pH has a bell-like shape, sin-
ce each enzyme is characterized by a pH range within which the rate of an enzyme-catalyzed reaction
attains a maximum. Any change in the pH value to either side leads to a decrease in the enzymic reac-
tion rate.
Optimal pH values for certain enzymes:
- pepsin = 1,5 2,5
- urease = 6,4 7,2
- trypsin = 7,5 7,8
- arginase = 9,5 9,9.
Dependence of enzymic reaction rate on temperature
When the temperature of medium is raised, the rate of an enzymic reaction increases, attains a
maximum at an optimal temperature, and then drops down to zero. The optimal temperature for most
enzymes falls within the range of 20-40
o
C. Thermal lability of enzymes is related to the proteinic
structure. Certain enzymes are denatured at temperatures above 40-50
o
C. A number of enzymes are
inactivated on cooling, i.e. their denaturation occurs at temperatures close to 0
o
C.
Estimation of enzymic activity: methods and units
The enzymes contained in cells, tissues, and organs must be preparative extracted by making use
of special techniques. The enzyme solution (extract from biological materials) is then used for enzyme
testing. Serum or blood plasma, as well as other biological fluids are ready enzyme solutions and can
be used for testing immediately. Enzyme tests, both qualitative and quantitative, are carried out in an
indirect manner by measuring a decrease in the substrate amount, or by estimating reaction products
accumulated in the medium.
The rate at which the substrate is consumed, or the reaction products are accumulated per unit
time, is taken as a measure of enzymic activity.
The determination is carried out using any suitable method (colorimetry, spectrophotometry, fluo-
rometry, polarography, etc.) either by recording the signal after the reaction is interrupted in a certain
period of time, or by taking measurements continually during the reaction. The latter technique is
more convenient, especially if the substrate or reaction products exhibit characteristic absorption in a
definite spectral region (the light absorption change in the course of the reaction is recorded by a spec-
trophotometer), or fluorescence (the fluorescence change is continually recorded within a certain time
interval using a spectrofluorimeter), etc. In other words, the choice of a method for estimating the
enzymic activity is primarily determined by the possibility of reliable assessment of the substrate and
reaction product concentrations.
Units of enzyme activity
According to the Nomenclature Committee of the International Union of Biochemistry, to specify
the activity of an enzyme, the use of the Unit (U) is recommended which is the amount of enzyme re-
quired to turn over one micromole of substrate per minute under standard conditions. Specific activity
of enzyme is expressed as the amount of enzyme (in milligrams) needed to consume one micromole
substrate per minute under standard conditions; its dimension is Mmol/minmg protein. A new unit of
catalytic activity, the katal (denoted kat), has also been recommended (1972), which expresses the
amount of enzyme required to turn over one mole substrate per second (mol/sec) under standard condi-
tions.
Regulation of enzyme activity
The enzymes are catalysts with controlled activity. Therefore, the rates of chemical reactions in
the organism can be monitored through the intermediacy of enzymes. The regulation of enzyme activi-
ty can be effected through the interaction of enzymes with various biological components or foreign
compounds (for example, pharmaceuticals or toxins) which are commonly called enzyme modifiers, or
enzyme regulators. The modifiers that are capable, by their action on enzymes, of accelerating reac-
tions are called activators; if the modifiers retard the reactions, they are referred to as inhibitors.
Activation of enzymes. The activation of an enzyme is defined by the acceleration of enzyme-
catalyzed reactions observable after the onset of modifier action. One group of activators is made up
of compounds affecting the active center region of an enzyme. This group includes substrates and en-
zyme cofactors. The cofactors (metal ions and coenzymes), while being essential structural elements
of conjugated enzymes act actually as cofactors for the latter. Metal ions, coenzymes, and substrates
22
(as precursors and active analogues of enzymes) can be used in practice as agents for activating the en-
zymes.
The activation of certain enzymes can be accomplished via structural modifications thereof, lea-
ving the active center of enzyme unaffected. A number of approaches to such a modification may be
envisaged:
1. The activation of an inactive precursor referred to as proenzyme, or zymogen.
2. The activation via addition of a specific modifying group to the enzyme molecule.
3. The activation via dissociation of an inactive complex protein-active enzyme.
Inhibition of enzymes. The investigation of enzymic inhibitory reactions is important from the
standpoint of practical applications for the synthesis of medicinal drugs, pesticides, etc., and in the elu-
cidation of the mechanisms of their action.
Still, a certain degree of caution should be exercised in employing the term inhibitor. In principle,
the inhibitor is understood as an agent capable of exerting a specific deterrent action on the activity of
an enzyme. The inhibitors are divided into two groups: reversible and irreversible inhibitors.
According to the mechanism of their action, the enzyme inhibitors are subdivided into the follo-
wing major types:
1. competitive
2. noncompetitive
3. uncompetitive
4. allosteric
5. substrate-linked.
Competitive inhibition is the enzymic reaction retardation produced by binding the enzyme acti-
ve center with an inhibitor structurally related to the substrate and capable of preventing the formation
of an enzyme-substrate complex. Under competitive inhibition conditions, the inhibitor and the sub-
strate, being structurally related species, complete for the active center of enzyme. The following sche-
me holds true for this type of inhibition:
E + I EI, where, I is the inhibitor, and EI, the enzyme-inhibitor complex.
The removal of inhibitory blocking can be accomplished by an excess of the substrate whose
molecules eliminate the inhibitor from the active center of the enzyme molecules and reactivate the
latter to catalytic activity. For example, malonic acid is a competitive inhibitor for succinate dehydro-
genase.
















Figure 10. Scheme of competitive and noncompetitive inhibition

Antimetabolites are promising for use as specific pharmaceuticals. Competitive relations are pos-
sible not only between the substrate and the inhibitor, but also between the inhibitor and the coenzy-
me.
Anticoenzymes (analogues of coenzymes, incapable of enzymic activity) likewise act as competi-
tive inhibitors by putting out of action the enzyme molecules to which they are bound. Anticoenzy-
mes (or their precursors, antivitamins) are widely used in biochemical studies and medicinal practice
as effective drugs.
23
Noncompetitive inhibition of enzymes is the retardation associated with the effect of an inhibi-
tor on the catalytic conversion rather than on the substrate-enzyme binding. A noncompetitive inhibi-
tor either directly binds the catalytic groups of the enzyme active center or, on binding with the enzy-
me, leaves the active center free and induces conformational changes in it. The conformational chan-
ges affect the structure of the catalytic site and hinder its interaction with the substrate. Since the non-
competitive inhibitor exhibits no effect on the substrate binding, in this case, as distinct from competi-
tive inhibition, formation of a ternary complex ESI according to the scheme below is observed:
E + S + I = ESI
However, no conversion of this complex to any reaction products occurs. Noncompetitive inhibi-
tors are exemplified by cyanides which are capable of strongly binding with the trivalent iron forming
part of the catalytic moiety of hemin enzyme, cytochrome oxidase. In intoxication, the toxin can be
bound or eliminated from the enzyme-inhibitor complex using reactivators, or antidots. These include
all SH-containing complexones (cysteine and dimercaptopropanol), citric acid, ethylenediaminetetra-
acetic acid (EDTA), etc.
Allosteric regulation of enzymic activity. The allosteric regulation is characteristic only of a
special group of enzymes with quaternary structure possessing regulatory centers for binding allosteric
effectors. The negative effectors, which retard the conversion of a substrate in the enzyme active cen-
ter, function as allosteric inhibitors.The positive effectors, on the contrary, accelerate enzymic reac-
tions and are therefore, assigned to allosteric activators. Most commonly, various metabolites, as well
as hormones, metal ions, and coenzymes act as allosteric effectors for enzymes. The greater the num-
ber of allosteric centers and effectors in an enzyme, the more is the enzyme responsive to metabolic al-
terations.
The mechanism of action of an allosteric inhibitor on enzyme is effected via a change of the en-
zymes active center conformation. Allosteric enzymes play an important role in the cell metabolism.
They take a key position in metabolism, since, being extremely responsive to metabolic change they
control the rate at which the materials are supplied through the whole enzymic system.
Multiple molecular forms of enzymes
A family of molecules might be envisaged for the same enzyme, and the term multiple enzyme
forms was coined to this effect. Commonly, a reference to multiple forms of enzyme implies the oc-
currence of enzyme proteins that differ among themselves in physico-chemical properties but can cata-
lyze the same reaction in the organism of a definite species. Depending on their origin, multiple enzy-
me forms are divided into two groups:
- isoenzymes (synonym isozymes),
- simply multiple forms of enzymes.














Figure 11. LDH-isoenzymes

Isoenzymes are molecular forms of an enzyme that arise due to genetic differences in the primary
structure of enzyme protein, i.e. distinctions in physico-chemical properties of isoenzymes are of gene-
tic origin. All nongenetically originated forms of the same enzyme are referred to as multiple enzyme
forms.
Isoenzymes of lactate dehydrogenase are hybrids of two subunits of independent genetic origin
(LDH
1
(4H), LDH
2
(3HM), LDH
3
(2H2M), LDH
4
(H3M) and LDH
5
(4M)). Isoenzymes of glutamate
24
dehydrogenase are polymers produced from subunits of the same type (different homopolymers)
nongenetic origination.
Multienzyme systems
The functioning of a multienzyme system is defined by the specificity of its cellular organization.
Types of multienzyme system organization may be envisaged: functional, structure-functional, and
combined types.
The functional organization is remarkable in that the individual enzymes are united in a function-
oriented multienzyme system through the agency of metabolites that are capable of diffusing from one
enzyme to another. In a functionally organized multienzyme system, the reaction product of the first
enzyme in the conversion chain serves as a substrate for the second enzyme, etc.
The structure-functional organization consists in that the enzymes form structural function-orien-
ted system via enzyme-enzyme (protein-protein) interactions. Such multienzyme complexes are tightly
bound and resist decomposition into constituent enzymes. This is their major distinction from functio-
nally organized multienzyme systems. For example, the enzymes involved may become fixed on the
biomembrane to form a chain. This is a pattern for the mitochondrial respiratory chain involved in
energy generation and transport of electrons and protons.

Polyenzymic complex Glycolytic enzymes


The combined type of multienzyme system organization is a combination of the two above types,
i.e. one part of the multienzyme system has a structural, and the other one, a functional organization.
This type of organization may be exemplified by the multienzyme system of the Krebs cycle in which
some of the enzymes are united into a structural complex (2-oxoglutarate dehydrogenase complex),
while other enzymes are functionally interrelated through metabolite mediators.
Immobilized enzymes
Immobilized, or insoluble, enzymes are an artificially derived complex of soluble enzymes bound
to a water-insoluble organic or inorganic carrier. The immobilization (from the Latin immobilis, fixed,
stable) is effected by:
- physical adsorption of an enzyme onto an insoluble material,
- entrapment of an enzyme in gel cells,
- covalent binding of an enzyme to an insoluble carrier,
- cross-linkage of enzyme molecules to form insoluble multienzyme complexes.
Glass, silica gel, hydroxylapatite, cellulose and its derivatives are commonly used as adsorbents.
However, immobilized enzymes are, as a rule, less active as compared with the free ones, since bin-
ding with the carrier weakens the enzyme-substrate contact. Insoluble enzymes can readily be remo-
ved from a reaction medium; they can be washed off the reaction products and repeatedly used in furt-
her experiments.
Practical utility of enzymes
Enzymes are widely used in practical activities of man. They are employed in various branches of
agriculture and technology, let alone their exceptional importance in medical practice.
In medicine the diagnostic importance of enzymes should be emphasized: the detection of indivi-
dual enzymes in clinical analyses is an aid in the identification of the nature of a disease. The enzymes
are used as substrates for a deficient enzyme in the organism, or as agents for the decomposition of a
25
substrate whose excess may be thought to be the cause of a presumed disease. Digestive enzymes
(pepsin, trypsin, etc.) are most commonly used in the clinic. Immobilized enzymes are used in the tec-
hnological syntheses of a number of hormonal preparations, in high-sensitive analyses of drugs, in
proximate analysis of biological components, and in many other applications. Proteolytic enzymes
(trypsin, chymotrypsin), immobilized on gauze bandages or tampons, are used in surgery for cleansing
purulent wounds and necrotic tissues; their action consists in enzymic degradation of dead cell pro-
teins discharged in purulent wounds. Currently, immobilized and soluble enzymes are most commonly
employed drugs of biological origin.

LECTURE 6

Subject: NUCLEIC ACIDS: STRUCTURE AND LEVELS OF NUCLEIC
ACIDS ORGANIZATION

Nucleic acids (nuclein from the Latin nucleus), or polynucleotides, are high- molecular com-
pounds composed of mononucleotides linked to chains by 3,5-phosphodiester bonds. The concentra-
tions of DNA and RNA in the cells are dependent on the functional state of the cells.
99% of DNA is present in nucleus, and 1% - in mitochondria. RNA, as distinct from DNA, is dis-
tributed more uniformly over the cell. In the cells of higher organisms, the nucleus accounts for about
11%, the mitochondria for about 15%, the ribosomes for 50 %, and the hyaloplasm for 24 % of the to-
tal of RNA. In the cell chromosomes of human and other higher organisms, the DNA is bound to his-
tones and nonhistone proteins. Such DNA-protein complexes are referred to as deoxyribonucleopro-
teins (DNP).
The molecular mass of RNA is significantly inferior to that of DNA. Depending on the function
assigned, molecular mass, and nucleotide composition, the following major types of RNA are distin-
guished: messenger RNA (m-RNA), transfer RNA (t-RNA), and ribosomal RNA (r-RNA). In the cell,
most RNA is bound to various proteins. Such complexes are called ribonucleoproteins (RNP).

Concise characterization of nucleic acids occurring in cells of higher organisms
DNA is storage of genetic information and mediation in transfer of its parent DNA in the course
of the cell division, or in transfer of RNA in the course of biological activity.
m-RNA is carrier of the DNA region containing structural information on protein polypeptide
chain. It participates in the information transfer from DNA (replication) to the site of protein synthesis
(on ribosome).
t-RNA participates in the activation of amino acids, their transport to ribosome, and assemblage
of polypeptides from amino acids on ribosome.
r-RNA forms of the skeleton of cytoplasmic ribosome (or mitochondria), to which ribosomal
proteins are linked in protein assemblage on ribosome.

Components of nucleic acids
Nitrogenous bases. By their chemical structure, the nitrogenous bases of nucleic acids are divi-
ded into two groups: purine bases and pyrimidine bases. Purine bases are adenine and guanine; pyri-
midine bases are cytosine, uracil, and thymine. DNA contains adenine, guanine, cytosine, and thymi-
ne. RNA contains the same bases excluding thymine, which is replaced by uracil.

adenine guanine cytosine


26

thymine uracil uracil tautomers

Figure 12. Structures of nitrogenous bases. Uracil tautomers: amide or lactam structure (left)
and imide or lactim structure (right)

A very important feature of these bases is their ability to undergo a lactam-lactim tautomerism.
For example, uracil.
Nucleosides. Compounds that contain nitrogenous bases linked to pentose are called nucleosides.
With reference to the pentose type involved, two different species of nucleosides are distinguished:
deoxyribonucleosides containing 2-deoxyribose, and ribonucleosides containing ribose.


thymidine uridine

Figure 13. Chemical structure of nucleosides

Nucleotides. Nucleotides are phosphoric acid esters of nucleosides. They are also divided into
ribonucleotides, which contain ribose, and deoxyribonucleotides, which contain 2-deoxyribose. The
phosphate group can add to the pentose ring at various positions: at positions 2, 3, and 5 in ribonu-
cleotides, and at positions 3 and 5 in deoxyribonucleotides. Free nucleotides, present in the cell, car-
ry a phosphate group in the 5 position. Nucleoside 5-phosphates are involved in the biological syn-
thesis of nucleic acids and are formed by decomposition of the latter. Since nucleoside 5-phosphates,
or mononucleotides, are derived from the corresponding nucleosides, major and rare ribomononucleo-
tides and deoxyribomononucleotides are distinguished.
The lengthening of the mononucleotide phosphate end by linking on additional phosphate group
to it leads to the formation of nucleoside polyphosphates:



Figure 14. Structural elements of the most common nucleotides

Nucleoside diphosphates and nucleoside triphosphates are those most frequently occurring in the
cells. The names and abbreviated notations for nucleoside phosphates are given below:
Name Notation
Ribonucleoside phosphates
Adenosine mono-, di-, triphosphate AMP, ADP, ATP
Guanosine mono-, di-, triphosphate GMP, GDP, GTP
Cytidine mono-, di-, triphosphate CMP, CDP, CTP
Uridine mono-, di-, triphosphate UMP, UDP, UTP

27

AMP

Deoxyribonucleoside phosphates
Deoxyadenosine mono-, di-, triphosphate dAMP, dADP, dATP
Deoxyguanosine mono-, di-, triphosphate dGMP, dGDP, dGTP
Deoxycytidine mono-, di-, triphosphate dCMP, dCDP, dCTP
Thymidine mono-, di-, triphosphate TMP, TDP, TTP
The entities ADP and ATP are macroenergetic, i.e. rich in energy; their chemical energy is used
by the organism for performing various functions. The other nucleoside diphosphates and triphospha-
tes are also implicated in the reactions of biological synthesis.

ATP
Nucleotide derivatives. Cyclic nucleotides (for example, 3,5-cAMP and 3,5-cGMP), which are
universal regulators of intracellular metabolism, merit special mention. A large number of nucleotide
derivatives serve as coenzymes for diverse enzymes in metabolic reactions. Coenzymes, derivatives of
mononucleotides and dinucleotides, are also known (FAD and FMN, NAD and NADP, etc., see Lec-
ture 4 Enzymes).

Cyclic adenosine monophosphate (3,5cAMP) and guanosine monophosphate (3,5cGMP)

Structure and levels of organization of nucleic acids
Nucleic acids possess primary, secondary, tertiary and quarternary levels of organization.
Primary structure of nucleic acids. The primary structure of DNA and RNA is linear polynu-
cleotide chain made up of mononucleotides which are linked by 3,5-phosphodiester bonds. The
build-up of both DNA and RNA primary structures follows the same basic principle: each pentose 3-
hydroxyl group of one mononucleotide is linked covalently to pentose 5-hydroxyl group of the neig-
hboring mononucleotide. Thence the name is derived: 3,5-phosphodiester bond. Linear chains of
DNA and RNA, whose lengths are determined by the number of constituent nucleotides, have two
ends called the 3-end and the 5-end.
Since nucleoside 5triphosphate is the starting material for assemblage of nucleic acids in the cell,
the 5-end of the chain contains a triphosphate, and the 3-end, a hydroxyl group. Consequently, the
chains of nucleic acids are polar and are directed either 53 or 35. Exceptions to the rule are
circular DNA and RNA of certain viruses and bacteria.
The secondary structure of DNA. In 1953, Watson and Crick proposed the double-helix model
for the DNA secondary structure. The DNA double helix is formed via specific pairing of a base of
28
one polynucleotide chain with a base of the other chain: the interaction of adenine with thymine is ef-
fected through involvement of two hydrogen bonds, and that of guanine with cytosine, through three
hydrogen bonds.
Such a mutual correspondence of the base pairs is called the complementarity. The DNA chains
are directed antiparallelly (one of the two chains runs in 53 direction, and the other one, in 35
direction). The sugar phosphate moieties of both chains are directed outwards, while the bases protru-
de into the interior of the helix, pair-wise against each other. The DNA helix is regular in structure:
one full turn of the helix contains 10 nucleotides and is 3.4 nm long. The length of each nucleotide in
axial direction is therefore 0.34 nm. Such a form of the Watson-Crick double helix is called the B-
form.




Figure 15. DNA secondary structure (from left to right: the structures of A, B and Z DNA)

The tertiary DNA structure emerges as the double-helical DNA molecule is twisted in space.
Outwardly, it looks like a super-coil or like a bent (broken) double helix. In higher organisms, the
DNA is located in chromosomes. Each chromosome contains a single giant DNA molecule of
molecular mass about 10
11
and a few centimeters long; this molecule constitutes a basis for chromatin.


Figure 16. Nucleosome structure

29
Chromatin is a super-molecular structure made up of a double-stranded DNA which is completed
primary with proteins and to a lesser extent with RNA and inorganic compounds. The major part of
chromatin is inactive. It contains a closely packed DNA. The active part accounts for 2-11% of the
total chromatin, depending on the nature of the cell. Chromatin resembles a string of beads, i.e.
globular bulges of about 10 nm in diameter spaced apart by thread-like linkers. The globular bulges
are called nucleosomes. Each nucleosome contains a length of double-stranded DNA equal in
extension to 140 base pairs, and eight molecules (an octet) of histones. The histone octet is composed
of four pairs of histones of H
2o
, H
2|
, H
3
and H
4
type. The thread-like linkers of double-stranded DNA
that is composed of 30-60 base pairs are bound with histone H
1
, each, the linker lengths being different
in various cells.
The quaternary structure of DNA is superspiralization of the tertiary structure of it (chromosome).
The secondary structure of RNA. RNA are single-stranded molecules, therefore their secondary
and tertiary structures are not regular.

Base pairing in RNA Ribose structure indicating numbering
of carbon atoms

mRNA is formed in the cell from a precursor, pre-mRNA. The pre-mRNA secondary structure
contains hairpins and linear regions. The secondary structure of mRNA is a bent chain; while the ter-
tiary structure looks like a thread wound round a spool. Functionally, the part of spool is played by a
special transport protein particle called an informofer.
tRNA. The secondary structure of tRNA has the shape of clover leaf. This structure is produ-
ced owing to intra-chain pairing of complementary nucleotides in certain regions of the tRNA chain.
Acceptor region (end or terminus) made up of four linearly linked nucleotides, three of which show
the same sequence, CCA, in all types of tRNA. The 3-OH hydroxyl of adenosine is free. The amino
acid bound to the adenosine 3-OH hydroxyl group is transported by tRNA to ribosomes, where the
protein synthesis takes place.
rRNA exhibits a secondary structure composed of helical regions which alternate with nonhelical
bent regions. The tertiary structure of rRNA constitutes a framework for the ribosome and is shaped as
a rod or a coil. Ribosomal proteins adhere to the tertiary structure on the outside.


Figure 17. Structures of RNAs: rRNA (left) and tRNA (right from yeast)

Physico-chemical properties of nucleic acids
30
Physico-chemical properties of nucleic acids are primarily determined by their high molecular
mass and by the level of structural organization. Specific features of nucleic acids show up in their:
- colloidal and osmotic properties,
- high viscosity and density of solutions,
- optical properties,
- aptitude to denaturation.
In solution, the molecules of nucleic acids exist as polyanions with distinctly pronounced acidic
properties.
Denaturation is a property inherent in the macromolecules possessing a spatial organization. De-
naturation is produced by heating and by the action of chemical agents which break hydrogen and van-
der-Waals bonds stabilizing the secondary and tertiary structures of nucleic acids. DNA regains its na-
tive double helix. This phenomenon is called renaturation.
Molecular hybridization of nucleic acids. The aptitude of nucleic acids to renaturate after dena-
turation has provided for a very valuable method of estimating the homology, or congenerity, of nucle-
ic acids. This method is referred to as the molecular hybridization. It is based on the complementary
pairing of single-stranded regions of nucleic acids.

LECTURE 7

Subject: TYPES OF GENETIC TRANSFORMATION TRANSFER.
MOLECULAR FUNDAMENTALS OF REPLICATION AND
TRANSCRIPTION

Three variants for genetic information transfer occurring in different organisms may be distingui-
shed.
1. Replication (or, less commonly, duplication) is the transfer of genetic information within a
single class of nucleic acids, i.e. from DNA to DNA or, as in certain viruses, from RNA to RNA.
2. Transcription is the transfer of information between different classes of nucleic acids, via
DNA-to-RNA scheme. As distinct from replication, transcription is effected as a transfer of a certain
portion of information stored in the DNA molecule. In the course of transcription, all the RNA types
(mRNA, tRNA, and rRNA) are formed. Distinguished are forward transcription (from DNA to RNA)
and reverse transcription (from RNA to DNA).
3. Translation is the transfer of genetic information from mRNA to a protein; it confined within
the macromolecules of different classes. The specified direction of genetic information transfer from
DNA via RNA to protein is called the central dogma of molecular genetics; it has been formulated by
Crick. According to the central dogma, no information transfer is allowed from protein to RNA. Ho-
wever, the RNA-to-DNA information transfer is not, in principle, forbidden. If, by any chance, infor-
mation transfer from protein to RNA is ascertained, this will require an immediate revision of the cur-
rently accepted basis of molecular genetics.
All the types of genetic information transfer are based on the template mechanism. This signifies
that each type requires an appropriate template. During replication, one of the two DNA chains (or
RNA in viruses) serves as a template. In transcription, a DNA section (forward transcription), or a
RNA section (reverse transcription), and in translation, mRNA, that is, only a nucleic acid is capable
of acting as a template. The precision of reproduction from the corresponding nucleic template is war-
ranted by the complimentary rule for the nitrogenous bases of nucleotides according to which the base
pairing occurs as A-to-T (or A-to-U in RNA) and G-to-C. Owing to this rule, the alternation order of
nucleotides in each new polynucleotide chain is complimentary to that of the template.

Molecular fundamentals of replication
In 1957, Meselson and Stahl established that DNA replication in living organisms proceeds by the
semiconservative mechanism. Untwisting proteins (helicase) break hydrogen bonds between the com-
plementary bases of the double-stranded DNA. As a result, the double helix becomes untwisted and
separates into single strands. Outwardly, this resembles the unzipping of a zipper. The untwisted
portion of DNA is called the replicative fork.
31
The initial step of replication (initiation) is the production of the RNA primer in the 53 direc-
tion, assisted by RNA-polymerase (primase). After the synthesis of the short chain of RNA on the
DNA template is completed, the enzyme is detached from DNA. Subsequently, deoxyribonucleotides



Figure 18. Replication of DNA: Scheme of the replication fork: a: template, b: leading strand,
c: lagging strand, d: replication fork, e: primer, f: Okazaki fragment

are added to the RNA primer through the assistance of DNA-polymerase III in 53 direction. A
hybrid chain RNA-DNA is thus formed.


















In this process, DNA-polymerase III synthesizes short DNA fragments (the so-called Okazaki
fragments) on the other parental strand of the replicative fork. An RNA primer is also needed for the
synthesis of Okazaki fragments. Interestingly, DNA-polymerase III is able, in the course of the synthe-
sis, to correct the errors in a mismatched pairing of nucleotides. If an error in base pairing occurs, the
mismatched nucleotide is immediately split off by the enzyme operative within the controlled nuclease
activity mechanism. At the same time, a correctly paired new nucleotide is routinely added, through
the agency of the enzyme, to the preformed DNA fragment.
The RNA primer, after the termination of DNA-polymerase III activity, is removed from the syn-
thetic chain either by specific ribonuclease H, or by DNA-polymerase I. At the site of the removed
RNA primer, the missing fragment of DNA strand is completed by means of DNA-polymerase I. The
colligation of the presynthesized DNA fragments (Okazaki fragments) in the 35 direction is effec-
ted through the aid of DNA-ligase.
Repair of DNA. The DNA repair, or correction of damaged sites in either of the DNA strands,
may be regarded as a limited replication. The repair process of DNA strands damaged by UV radiation
has been studied in greater detail (for example, radiation damage of epithelial cells of the skin).
UV radiation causes cross-links between the neighboring thymine residues in DNA. These di-
merized thymine residues are thus rendered incapable to perform the template function during replica-
tion. To provide against this contingency, the cell has a complement of enzymes whose function is to
repair damage inflicted on DNA. At first, the damaged spot is recognized and excised by DNA-ases.
Then the correct sequence in the 53direction is synthesized by a DNA-polymerase I enzyme type.
32
Finally, the ends of the new section are joined to the former strand by DNA-ligase. Inherited mutations
that affect DNA repair genes are strongly associated with high cancer risks in humans.




DNA repair rate is an important determinant of cell pathology

Molecular fundamentals of transcription
During the biological activity of the mature cell, only a portion of the genetic information that has
been written down in the chromatin DNA is used in the transcription as RNA copies. In procaryotes
and eucaryotes, the elementary transcription unit, i.e. a DNA segment liable to transcription, is called
transcription. The section is the structural gene, that carry information are called exons, while those
that carry none, introns. Possibly, the introns perform a supplementary regulatory function for the
exons. In the chromosomal DNA, mobile genes, or transposons, have been identified.
Mechanism of transcription. The starting point of transcription is called a promotor. Proteins
that facilitate the transcription initiation and the transcription enzyme RNA polymerase are added to
the promotor from the medium. The operator is the section of DNA capable of binding proteins that
act as transcription regulators. In procaryotes, the transcription regulator protein is the repressor. In eu-
caryotic DNA, the transcripton section adjacent to the promotor is called the acceptor, or regulatory
site. At the transcript on end, there is a nucleotide sequence whose function is to signal the termination
of transcription, the so-called terminator. The RNA produced by transcription is called the transcript.
The transcript is a complementary copy of transcript on extending from the promotor to the terminator.
The transcription of DNA may be considered as proceeding via three stages: initiation, elonga-
tion, and termination of RNA synthesis.
The initiation of transcription occurs as DNA-dependent RNA-polymerase becomes bound to
the DNA promotor exhibiting a high affinity for this enzyme. The promotor is the starting site of trans-
cription. The procaryotic RNA-polymerase is composed of five different subunits. Four of these cons-
titute an aggregate called the core enzyme whose function is to catalyze the formation of phosphodies-
ter bonds between RNA nucleotides. The fifth subunit called sigma (o)-factor, is readily detachable
from the core enzyme. This o-factor acts to select the starting site for the transcription by binding with
the promotor. The transcription site thus having been specified, the core enzyme is added to the o-fac-
tor to initiate the transcription. It remains unclear as yet what makes the strands of double-helix DNA
separate at the transcription site. Probably this function is performed by RNA-polymerase itself, or, al-
ternatively, there is a protein specially intended for this purpose (like untwisting proteins in repara-
tion).
Eucaryotes have RNA-polymerases of three types: I, II, and III. These are proteins composed of
several subunits and differing in transcription specificity. RNA-polymerase I is responsible for the
transcription of rRNA genes, RNA-polymerase III, for the transcription of tRNA, and RNA-polymera-
se II participates in the synthesis of a precursor to mRNA. RNA-polymerases make the chain grow in
the 53 direction only; for this reason, the 5-end always bears a triphosphate (P~P~P), and the 3-
end, a free hydroxyl, OH.
The elongation (or polymerization) of transcription proceeds as RNA-polymerase move along
the DNA template. Every next nucleotide becomes paired to a complementary base in the codogenic
33
DNA template, and RNA-polymerase fastens the nucleotide through phosphodiester bonds to the
growing RNA chain.
The elongation rate is about 40 to 50 nucleotides per second. Within 1 second after the transcrip-
tion onset the nascent transcript is about 30 nucleotides long, a protective chemical group, the so-cal-
led cap, is formed at its 5-end. In other words, simultaneously two different acts of transcript gro-
wth are operative: the transcription proper and the post-transcriptional modification of transcript
(capping). The cap is present in all eucaryotic mRNA. It is composed of 7-methylguanosine linked
through a chain of three phosphate groups and 55, rather than 53, phosphodiester bond to the
first mRNA nucleotide, CH
3
-G-P-P-P-mRNA. This methylated cap protects the mRNA from an attack
by 5-exonucleases. Length of primary transcript is 5000-8000 nucleotides, but it may be long as
20,000 and even more.
The termination of transcription takes place as RNA-polymerase has reached the DNA nucleoti-
de sequences, called termination codons, which signal to stop the polymerization. It is believed that
such termination codons in a transcription may be poly (A) sequences, since in transcripts at the 3-
end, the corresponding complementary poly(U) sequences occur. A special termination factor, rho ()-
factor, which is a protein, has been identified. The -factor interrupts the transcription by recognizing
in a specific, as yet not quite clear, manner the transcription termination codons. Owing to the
terminators, the RNA chains of a definite length only are formed.
As the transcription proceeds to its termination, the synthesized RNA separates itself from the co-
dogenic DNA strand. The primary products of transcription, i.e. RNAs, are complete copies (in com-
plementary representation) of the DNA transcriptons. This implies that the newly synthesized RNA
contains both informative and noninformative portions. The primary transcript is also called the RNA
precursor. The mRNA precursor, or pre-mRNA, is represented by linear chain that does not close to a
ring. It is longer as compared with functionally active RNA molecules. Pre-mRNA is especially liable
to variation in the molecular mass: from 6-7 S to giant sizes of 50-70 S. In eucaryotic nuclei, all the
precursors are bound to proteins to form ribonucleoproteins.

Post-transcriptional alterations in RNA
In the nucleus, all the RNA precursors are involved in the stage of post-transcriptional matura-
tion, or processing. During processing, all noninformative, redundant fragments are removed from
the pre-RNA to form mature, functionally adequate RNA molecules. Processing includes three
steps:
1. Excision of noninformative portions from pre-RNA.
2. Splicing of the informative portions of the split genes.
3. Modification of the 5- and 3-ends in RNA.
Pre-mRNA processing. In the course of processing, the modified primary transcript is excised at
the intron sites, and the exons are spliced by their ends to form mRNA. This function is carried out by
the so-called small nuclear ribonucleoproteins, rich in uracil. One of these, denoted U
1
-RNA, is
believed to play a major role in excising the introns and in splicing the exons, i.e. this molecule
possesses a function of both ribonuclease and ligase. The introns are bent to form a loop, which
facilitates their excision, while the exon ends become linked through phosphodiester bonds.


Figure 19. Splising of mRNA

In the nucleus, the 5- and 3-ends of the mRNA formed are modified. A polyadenyl fragment,
poly (A), composed of about 200 nucleotides, is added to the 3-end of eucaryotic mRNA. This addi-
tion is accomplished by means of poly (A)-polymerase. The poly (A) fragment is apparently needed
for the transport of mRNA from nucleus to cytoplasm.
Transport of mature RNAs from the nucleus to the cytoplasm. Eucaryotes, in distinction from
procaryotes, possess a nucleus membrane, and the ready RNA must pass across the membrane to be
34
supplied to the cytoplasm where protein synthesis occurs. All the mature RNAs are transported from
the nucleus into the cytoplasm as complexes with proteins; the proteins facilitate the transport and pro-
vide an additional protection from RNA environmental factors. The mRNA becomes bound to a spe-
cial protein, informofer (which means information carrier). RNA, complexed with the informofer, is
supplied to the cytoplasmic ribosomes, within which the assembly of proteins from amino acids, or
transcription, takes place.
Reverse transcription. In the fields of molecular biology and biochemistry, a reverse
transcriptase, also known as RNA-dependent DNA polymerase, is a DNA polymerase
enzyme that transcribes single-stranded RNA into double-stranded DNA. It also helps in the
formation of a double helix DNA once the RNA has been reverse transcribed into a single
strand cDNA. Normal transcription involves the synthesis of RNA from DNA; hence, reverse
transcription is the reverse of this. Well studied reverse transcriptases include:
- HIV-1 reverse transcriptase from human immunodeficiency virus type 1 (PDB 1HMV).
- M-MLV reverse transcriptase from the Moloney murine leukemia virus.
- AMV reverse transcriptase from the avian myeloblastosis virus.
- Telomerase reverse transcriptase that maintains the telomeres of eukaryotic chromosomes.
The enzyme is encoded and used by reverse-transcribing viruses, which use the enzyme during
the process of replication. Reverse-transcribing RNA viruses, such as retroviruses, use the enzyme to
reverse-transcribe their RNA genomes into DNA, which is then integrated into the host genome and
replicated along with it. Reverse-transcribing DNA viruses, such as the hepadnaviruses, can allow
RNA to serve as a template in assembling, and making DNA strands. HIV infects humans with the use
of this enzyme. Without reverse transcriptase, the viral genome would not be able to incorporate into
the host cell, resulting in the failure of the ability to replicate.
Process of reverse transcription. Reverse transcriptase creates single stranded DNA from an
RNA template. In virus species with reverse transcriptase lacking DNA-dependent DNA polymerase
activity, creation of double-stranded DNA can possibly be done by host-encoded DNA polymerase ,
mistaking the viral DNA-RNA for a primer and synthesizing a double-stranded DNA by similar me-
chanism as in primer removal, where the newly synthesized DNA displaces the original RNA templa-
te.The process of reverse transcription is extremely error-prone and it is during this step that mutations
may occur. Such mutations may cause drug resistance.
I n eukaryotes. Self-replicating stretches of eukaryotic genomes known as retrotransposons utili-
ze reverse transcriptase to move from one position in the genome to another via a RNA intermediate.
They are found abundantly in the genomes of plants and animals. Telomerase is another reverse trans-
criptase found in many eukaryotes, including humans, which carries its own RNA template; this RNA
is used as a template for DNA replication.




Scheme of reverse transcription
Antiviral drugs. As HIV uses reverse transcriptase to copy its genetic material and generate new
viruses (part of a retrovirus proliferation circle), specific drugs have been designed to disrupt the process
and thereby suppress its growth. Collectively, these drugs are known as reverse transcriptase inhibitors
35
and include the nucleoside and nucleotide analogues zidovudine (trade name Retrovir), lamivudine
(Epivir) and tenofovir (Viread), as well as non-nucleoside inhibitors, such as nevirapine (Viramune).

The molecular structure of zidovudine (AZT) used to inhibit HIV reverse transcriptase

Telomerase is an enzyme that adds DNA sequence repeats ("TTAGGG" in all vertebrates) to the
3' end of DNA strands in the telomere regions, which are found at the ends of eukaryotic chromoso-
mes. This region of repeated nucleotide repeats called telomeres contains non-coding DNA material
and prevents constant loss of important DNA from chromosome ends. As a result, every time the chro-
mosome is copied only 100-200 nucleotides are lost, which causes no damage to the organism's DNA.
Telomerase is a reverse transcriptase that carries its own RNA molecule, which is used as a template
when it elongates Carol W. Greider and Elizabeth Blackburn in 1984 in the ciliate Tetrahymena.
Together with Jack W. Szostak, Greider and Blackburn were awarded the 2009 Nobel Prize in
Physiology or Medicine for their discovery.
The protein composition of human telomerase was identified in 2007 by Scott Cohen and his
team at the Children's Medical Research Institute in Australia. It consists of two molecules each of hu-
man telomerase reverse transcriptase (TERT), telomerase RNA (TR or TERC), and dyskerin (DKC1).
The genes of telomerase subunits, which are TERT, TERC, DKC1, and TEP1 etc, are located on the
different chromosomes in human telomeres, which are shortened after each replication cycle. The
existence of a compensatory shortening of telomere (telomerase) mechanism was first predicted by
Soviet biologist Alexey Olovnikov in 1973, who also suggested the telomere hypothesis of aging and
the telomere's connections to cancer. Telomerase was discovered by genome. Human TERT gene
(hTERT) is translated into a protein of 1132 amino acids. TERT proteins from many eukaryotes have
been sequenced. TERT polypeptide folds with TERC, a non-coding RNA (451 nucleotides long in
human). TERT has a 'mitten' structure that allows it to wrap around the chromosome to add single-
stranded telomere repeats.
TERT is a reverse transcriptase, which is a class of enzyme that creates single-stranded DNA
using single-stranded RNA as a template. Enzymes of this class (not TERT specifically, but the ones
isolated from viruses) are utilized by scientists in the molecular biological process of reverse transcrip-
tase PCR (RT-PCR), which allows the creation of several DNA copies of a target sequence using
RNA as a template. As stated above, TERT carries its own template around, TERC.
The high-resolution protein structure of the Tribolium castaneum catalytic subunit of telomerase
TERT was decoded in 2008 by Emmanuel Skordalakes and his team at The Wistar Institute in Phila-
delphia. The structure revealed that the protein consists of four conserved domains (RNA-Binding Do-
main (TRBD), fingers, palm and thumb), organized into a ring configuration that shares common fea-
tures with retroviral reverse transcriptases, viral RNA polymerases and bacteriophage B-family DNA
polymerases.
By using TERC, TERT can add a six-nucleotide repeating sequence, 5'-TTAGGG (in all verte-
brates, the sequence differs in other organisms) to the 3' strand of chromosomes. These TTAGGG re-
peats (with their various protein binding partners) are called telomeres. The template region of TERC
is 3'-CAAUCCCAAUC-5'. This way, telomerase can bind the first few nucleotides of the template to
the last telomere sequence on the chromosome, add a new telomere repeat (5'-GGTTAG-3') sequence,
let go, realign the new 3'-end of telomere to the template, and repeat the process.
Aging. The enzyme telomerase allows for replacement of short bits of DNA known as telomeres,
which are otherwise shortened when a cell divides via mitosis.
In normal circumstances, without the presence of telomerase, if a cell divides recursively, at some
point all the progeny will reach their Hayflick limit. With the presence of telomerase, each dividing
cell can replace the lost bit of DNA, and any single cell can then divide unbounded. While this unbou-
36
nded growth property has excited many researchers, caution is warranted in exploiting this property, as
exactly this same unbounded growth is a crucial step in enabling cancerous growth.
Embryonic stem cells express telomerase, which allows them to divide repeatedly and form the
individual. In adults, telomerase is highly expressed in cells that need to divide regularly (e.g., in the
immune system), whereas most somatic cells express it only at very low levels in a cell-cycle-depen-
dent manner.
A variety of premature aging syndromes are associated with short telomeres. These include Wer-
ner syndrome, Ataxia telangiectasia, Ataxia-telangiectasia like disorder, Bloom syndrome, Fanconi
anemia and Nijmegen breakage syndrome. The genes that have been mutated in these diseases all have
roles in the repair of DNA damage, and their precise roles in maintaining telomere length are an active
area of investigation. While it is currently unknown to what extent telomere erosion contributes to the
normal aging process, maintenance of DNA in general and telomeric DNA, to be specific, have eme-
rged as major players. Dr. Michael Fossel has suggested in an interview that telomerase therapies may
be used not only to combat cancer but also to actually get around human aging and extend lifespan sig-
nificantly. He believes human trials of telomerase-based therapies for extending lifespan will occur
within the next 10 years. This timeline is significant because it coincides with the retirement of Baby
Boomers in the United States and Europe.
Some experiments have raised questions on whether telomerase can be used as an anti-aging the-
rapy, namely, the fact that mice with elevated levels of telomerase have higher cancer incidence and
hence do not live longer. In addition, although certain premature aging syndromes have been associa-
ted with telomere shortening, mice without active telomerase do not appear to suffer from premature
aging. Telomerase also favors tumorogenesis, leading to questions about its potential as an anti-aging
therapy. On the other hand, one study showed that activating telomerase in cancer-resistant mice by
overexpressing its catalytic subunit extended lifespan. The potential remains for telomerase activators
to contribute to the development of cancer.http://en.wikipedia.org/wiki/Telomerase -
cite_note-Nobel_Prize_2003-2#cite_note-Nobel_Prize_2003-2

LECTURE 8

Subject: MOLECULAR PRINCIPLES OF TRANSLATION

During translation, the mRNA genetic text is translated into a linear sequence of amino acids that
constitute the polypeptide chain of a protein. Since the product of translation is a specific protein, the
translation process may with equal right be called protein biosynthesis.

Genetic code and its characterization
The genetic, or amino acid, code is understood as the relationship between the sequence of bases
in a nucleic acid and the sequence of amino acids in the polypeptide synthesized from it; otherwise
stated, this is the correspondence of codons (code words) to definite amino acids. The genetic code
may be regarded as a specific dictionary for translating a text, recorded by means of four nucleotides,
into a protein text, recorded by means of 20 amino acids. The other amino acids found in a protein are
modified forms of one of the 20 amino acids. The genetic code exhibits the following features:
1. Triplicity: a triplet of nucleotides corresponds to each amino acid. There are four nucleotides
available; it can easily be seen that, taken as triplets, they can form 4
3
=64 codons. Of these, 61 are
sense codons, and 3, nonsense codons (or termination codons).
2. Nonoverlap: The genetic text codons are independent of each other.

|CCA| |CGG| |AAC|
Nonoverlapping

3. Degeneracy, or redundancy: certain amino acids may have more than one codons. A simple
comparison testifies to this: 61 sense codons account for 20 amino acids, i.e. on the average, are sligh-
tly more than 3 codons per amino acid.
4. Specificity: definite codons correspond to each amino acid. They cannot be used for another
amino acid.
5. Colinearity: correspondence between the linear sequence of codons in mRNA and that of ami-
no acids in protein.
37
6. Universality: all the above mentioned features of the genetic code are characteristic of any
living organism.

Mechanism of translation process
The translation process may be divided into two stages:
- the recognition of amino acids
- protein biosynthesis in its proper sense.
The spatial localization of these stages in the cell is different: recognition occurs in the hyalo-
plasm, and protein biosynthesis proceeds on ribosome.
Recognition of amino acids. In essence, the amino acid recognition process consists in binding
an amino acid to its tRNA. The tRNA structure exhibits the properties of a potential translator, since
the tRNA is capable of both reading a nucleotide text (the tRNA anticodon is specifically paired
with the mRNA codon) and carrying its amino acid (at the acceptor end). However, the tRNA is inca-
pable of linking with its amino acid. For this purpose, there are available special enzymes in the cell
sap (cytosol) that actually act as translators, i.e. provide for the conditions enabling the synthetases
(for short, ARSases). At least 20 types of ARSases are known (by the number of proteinogenic amino
acids involved). They recognize specifically tRNA and its amino acid, and catalyze their addition ac-
cording to the reaction:

R-CH-COOH + HO-tRNA + Mg
2+
+ ATP ARSase
|
NH
2

R-CH-C=O
| O-tRNA + AMP + H
4
P
2
O
7

NH
2

ATP is required for this process, with Mg
2+
acting as a cofactor. The ATP energy is supplied to
the production of a macroergic bond in the aminoacyl-tRNA, i.e. the reaction simultaneously involves
the activation of amino acid at the carboxyl end and the addition of amino acid to the adenosine hydro-
xyl (3-OH) at the acceptor end (CCA) of tRNA. The cell contains about 40 to 60, rather than 20,
tRNAs, since some of the amino acids stand in need of specifically using more than one tRNA.
Further, the tRNA, with the amino acid linked to it, is transferred by simple diffusion to the ri-
bosome. The ribosomes perform the assembly of proteins from amino acids supplied as a variety of
aminoacyl-tRNAs.
Ribosomal protein biosynthesis. Protein biosynthesis (second stage of translation) requires:
- mRNA as a genetic template whose program defines the amino acid sequence order in a given
protein;
- aminoacyl-tRNA (for reading the genetic mRNA text, as a source of amino acids for the pro-
tein assembly);
- ribosomes as molecular assembly units for the successive linking of amino acids into a poly-
peptide chain in accordance with the mRNA program;
- GTP as an energy source in the ribosomal protein synthesis;
- protein factors assisting at various steps of the ribosomal protein synthesis;
- certain metal ions for using as cofactors (Mg
2+
, K
+
, and others).
Mechanism of ribosomal protein synthesis. Protein biosynthesis, or translation, proper is con-
ventionally divided into three stages:
1. initiation (start of synthesis),
2. elongation (polypeptide chain lengthening),
3. termination (end of synthesis).
Initiation. The starting stage is the slowest one in the overall translation process. In an
Inoperative stage, the ribosomal subunits are separated. The mRNA, transported from the nucleus into
the cytoplasm becomes bound to a small subunit at the site in its surface shared with the large subunit.
To be noted, the mRNA binding site is always located close to the 5-end of mRNA, since the
RNA program reading always proceeds, in the 53 direction. Within the subunit limits only two
mRNA codons can be accommodated. The first mRNA codon at the 5-end is AUG or GUG. These
codons are called initiation codons (or start codons), since they always serve as the start signal for the
ribosomal translation. In the tRNA, an anticodon of methionyl-tRNA is correspondent to these codons.
Eucaryotic cells have two different methionyl-tRNAs. One of the two is always involved in the initia-
38
tion, while the other one is engaged in the elongation process. In procaryotes, protein biosynthesis
starts from formylmethionyl-tRNA in which the NH
2
group is blocked by formyl moiety.



Figure 20. Diagram showing the translation of mRNA and the synthesis of proteins
by a ribosome

In addition, at least three proteinic initiation factors (IF
1
, IF
2
, and IF
3
), which are not ribosomal
constituents, and GTP take part in the initiation. The initiation factors facilitate the binding of mRNA
with the small subunit (S) and GTP. A large subunit is added to the primary complex thus formed (IF-
S-mRNA-GTP) to complete the closure of all the ribosomal subunits; this having been performed, the
initiation factors are removed from the ribosome. The energy needed for the ribosomal subunit closure
is supplied by GTP hydrolysis. The initiator complex thus formed (mRNA-ribosome-methionyl-
tRNA) is ready to start elongation. To be noted, methionyl-tRNA, through its anticodon, becomes spe-
cifically paired to the AUG codon of mRNA, i.e. it becomes as if suspended from the mRNA by
hydrogen bonds, while its acceptor site (for binding an amino acid) is attached to the large ribosomal
subunit.
Elongation. The polypeptide synthesis always starts from the N-end and terminates in the C-end.
The addition of one amino acid to the polypeptide chain is accomplished in three steps:
- binding of aminoacyl-tRNA
- transpeptidation (or peptide transfer)
- translocation (or displacement of mRNA by one triplet).
The first step. In the ribosome tRNA is located on the right; tRNA is bound to the mRNA codon
through its anticodon, while its acceptor end is linked to the growing peptide. This peptide, which is
a constituent of peptidyl-tRNA, is bound to the P-site(peptide, or donor site), which is a kind of pro-
tein pocket in the large subunit. As the first step is being performed, the second mRNA codon is free.
This codon becomes coupled to the anticodon of the incoming aminoacyl-tRNA. The aminoacyl end
of tRNA becomes bound to the A-site (acceptor site) of the large ribosomal subunit. This brings to ter-
mination the first step, i.e. binding. Energy for the binding is provided by breaking the GTP phosphate
bond.
The second step, transpeptidation, proceeds in such a manner that peptidyl is transferred from the
left-side tRNA onto the amino group of aminoacyl-tRNA. A peptide bond buildup is catalyzed by
ribosomal proteins exhibiting a peptidyl transferase activity.
The third step involves separation of the ribosomal subunits using the energy supplied by one
GTP molecule. Peptidyl-tRNA that carries the tripeptide moves, together with the mRNA to which it
is coupled, a distance of one codon from the A-site to be positioned at the P-site. This movement is
called translocation; it is accompanied by the release of free tRNA from the ribosome. For the synthe-
sis of one peptide bond (or lengthening of the polypeptide by one amino acid), the energy of two GTP
molecules is consumed.
Elongation is assisted by the so-called elongation factors (or transfer factors), proteins catalyzing
the elongation of peptide chains. Elongation continues until all the mRNA text has been completely
read out.
Termination, or the completion of polypeptide synthesis, is dependent on the occurrence of ter-
mination codons, or stop-signals (UAA, UGA, UAG), and elongation factors in mRNA. The tRNAs
39
are not capable of binding with termination codons, since they are not in possession of the correspon-
ding anticodons. It is possible that the termination factors release the synthesized polypeptide chain. In
the cell mRNA uses more than one ribosome for protein biosynthesis purposes. Such along the length
of a strand of mRNA is called polyribosome. The polypeptide chain is lengthened at a rate of one ami-
no acid per second; and during an intensive phase of cell growth, the biosynthesis rate rises to 20 ami-
no acids per second.

Post-translational modifications of proteins
During translation, the protein starts to be folded into a three-dimensional structure which is com-
pleted to the final form after the protein has been released from the ribosome. Some proteins are syn-
thesized as precursors. These are subject to a limited hydrolysis in the cell. Apparently, most proteins
are amenable to the treatment by proteases, i.e. figuratively speaking they pass a stage of maturation.
A significant part of the synthesized proteins remain inside the cell, while some proteins are parti-
cular active in protein secretion. As a rule, the proteins that have been synthesized on ribosome bound
with the endoplasmic reticulum (EPR) membrane are exported from the cell. In the cisterns of endo-
plasmic reticulum, the proteins become concentrated. The storage and secretion of proteins take place
in the Golgi apparatus, where a carbohydrate component is added to the proteins. The transport of
proteins from the cell is accomplished by exocytosis. The ATP energy is consumed in this process;
when ATP is in short supply, the proteins are retained within the cell.
Conservation of mRNA. The mRNA intended for storage and subsequent re-usage of its genetic
program for assembling the required proteins must be protected against an attack by nucleases. In the
cell, the conservation of mRNA is done by binding it to special cytoplasmic proteins. Such protein-
mRNA arises for protein biosynthesis, mRNA binding to the small ribosomal subunit to be engaged in
the translation. The mRNA conservation procedure is used during cell development.



Figure 21. Post translation modification of protein (folding of protein: from primary
to quaternary structure)

Regulation of protein biosynthesis
Proteins determine the vital activity of the cell. Therefore, within the framework of the overall
scheme of cellular protein synthesis, the cell must exercise a precise control over the synthesis of the
proteins needed at the given moment.
40
Proteins that are synthesized at a constant rate are called constitutive proteins, while those for
which the synthesis rate may be varied within a wide range depending on the extant conditions are cal-
led adaptive, or inducible, proteins. The concentrations of inducible (adaptive) proteins are subject to
wide variation. The synthesis of constitutive proteins appears to be independent control, and in con-
trast, the synthesis of inducible proteins is liable to a very strict regulation.











Regulation of protein biosynthesis (A repression, B induction)

The stimulation of protein biosynthesis, which leads to an increased amount of proteins, is called
induction, while the inhibition of protein biosynthesis is referred to as repression. Apparently, there
are substances in the cell that are capable of signaling the state of metabolism in the cell or in the orga-
nism. This allows switching protein synthesis on or off. In procaryotes, such signal compounds may be
nutrients supplied to the cell, metabolites, and certain intracellular regulators (of cyclic nucleotide ty-
pe). In multicellular systems, especially highly organized ones, in addition to autonomous intracellular
regulators, an important role is given to extracellular protein biosynthesis regulators which govern the
activity of the genetic apparatus of protein biosynthesis for a given tissue or organ cell in accordance
with the global strategy of the whole organism.

Genetic engineering
Genetic engineering is a branch of molecular genetics dealing with the development of methods
for constructing desired genes and incorporating them into the host cell with the purpose of changing
the genetic properties of the cell. In future methods of genetic engineering have proved to be effective
in the transplantation of numerous genes, including those of insulin, somatotropin, ovalbumin, and ot-
hers. This opens the way for the potential commercial production of proteinic drugs by the genetic en-
gineering method.

Preparations affecting protein biosynthesis
Preparations that affect protein biosynthesis are widely used in practice. Inducers are applied to
stimulate protein synthesis in impaired cells, or in the organs affected by long-lasting inactivity (atro-
phied organs). This inducer effect facilitates the functional restoration of the cells in a damaged organ.
Protein synthesis inhibitors are used for the opposite effect, i.e. to suppress the division and growth of
cells.
Preparations for stimulating protein synthesis belong to the so-called anabolic agents. Hormonal
and nonhormonal anabolic agents are distinguished. The group of hormonal agents is the larger one.
Among these agents, the most pronounced ability to protein synthesis induction (at the transcription
level) is manifest in the anabolic steroids (methandrostenolone, phenobolin, and the most active reta-
bolil) which are derivatives of male sex hormones (androgens) and are used with the exclusive purpo-
se to stimulate protein synthesis in the organism. Insulin exhibits a distinct anabolic activity; this pro-
tein hormone appears to activate protein biosynthesis at the translation level.
Nonhormonal anabolic agents that have gained acceptance in practice include precursors of nuc-
leotides and nucleic acids. For example, sodium orotate (orotic acid is a key compound in the biosyn-
thesis of pyrimidine nucleotides) and inosine (or hypoxantine riboside).
Protein synthesis inhibitors. These constitute a larger group of preparations used in biochemical
studies and practical medicine. All the protein synthesis inhibitors may be classified into:
- transcription inhibitors
- processing and RNA transport inhibitors
- translation inhibitors.
41
The transcription inhibitors are: o-amanitin (poison of the death cup fungus, Amanita phalloi-
des), antibiotics rifamycins, actinomycin D (used in biochemical experiments), antibiotics olivomycin
and dactinomycin, plant alkaloids vinblastine and vincristine (used in medicine as antitumoral agents),
5-fluorouracil.

Processing and m-RNA transport inhibitors
Cordycepin (3-deoxyadenosine) may be referred to as an mRNA transport inhibitor since it im-
pedes the addition of polyadenyl moiety to mRNA, the polyadenyl moiety facilitating the transport of
mRNA from the nucleus to cytoplasm.
Translation inhibitors may be exemplified by antibiotics used as antibacterial agents in medi-
cine: chloramphenicol, lincomycin, erythromycin, tetracyclines, streptomycin, etc.

LECTURE 9

Subject: GENERAL CHARACTERIZATION OF METABOLISM AND
ENERGY METABOLISM

Characterization of metabolism
The vital activity of any living organism is determined by the specific organization of biological
structures, metabolic processes, energy metabolic processes, energy metabolism, genetic information
transfer, and regulatory mechanism. Damage of any of these links develops a pathological process and
a disease in the organism. An understanding of the molecular mechanisms involved in the vital activity
or malfunction of the organism constitutes the basis for the search and clinical applications of biologi-
cal medicinal preparations.
In the metabolism of the living organism distinguished are:
- exogenous metabolism, which comprises extra-cellular transformations of materials on the way
to their uptake and excretion by the cells
- intermediary metabolism, which occurs in the cells.
The intermediary metabolism is conceived as the total sum of chemical reactions that occur in the
living cell.
Functionally, metabolism encompasses the following major processes:
1. Accumulation of energy from decomposition of compounds or supplied by light.
2. Utilization of energy for synthesis of essential molecular components (monomers, macromole-
cules) and the performance of work (osmotic, electric, mechanical).
3. Decomposition of renewable structural components of the cell.
4. Synthesis and decomposition of specialized biological molecules (hormones, mediators, hor-
monoids, cofactors, etc.).
The sequences of chemical reactions involved form metabolic pathways, or cycles, each of these
performing a definite function. Conventionally, central and special metabolic pathways are distingui-
shed. Central pathways are common to the decomposition and synthesis of major macromolecules.
Actually, they are much alike in all representatives of the living world. Special cycles are characteris-
tic of the synthesis and decomposition of individual monomers, macromolecules, cofactors, etc. Spe-
cial cycles are extremely diversified.
In the metabolism, two oppositely directed processes, or phases are commonly distinguished:
catabolism and anabolism.
42


Catabolism of substances (3 stages)
Catabolismis the sum of degradation processes leading to the cleavage of large molecules into
smaller ones. Catabolism is accompanied by a release of energy that can be stored as energy-rich ATP.
Anabolism is the sum of metabolic processes leading to the synthesis of complex molecules from
simpler ones. Anabolic processes proceed through consumption of ATP and decomposition of the lat-
ter into ADP and H
3
PO
4
.
However, ATP is not the only linking component shared by catabolism and anabolism. Other
simple metabolites are also formed by the catabolic pathway from macromolecules and monomers to
be used as starting materials for the subsequent synthesis of monomers and macromolecules, i.e. in the
process of anabolism. This linking pathway, or cycle, unifying degradation and synthetic routes, is cal-
led the amphibolic pathway. This signifies that the catabolic and anabolic pathways are coupled not
only via the energetic ATP-ADP system, but also through their common metabolites, which renders
the metabolism more versatile and economical (for example, pyruvate, oxaloacetate, -ketoglutarate,
acetyl-ScoA, etc.).

Energy transfer in biochemical processes
The biological activity of the cell is closely associated with the continuous redistribution of the
energy delivered by the compounds that enter the cell. The storage of energy in the specific phosphate
bonds of ATP constitutes the basis for the energy transfer mechanism in the living cell. The living cell
is a nonequilibrated chemical system the circumstance that permits storing the energy, produced by
catabolic reactions of nutrients, in the ATP phosphate bonds.
The ATP energy in the cell can be converted, via three major routes, to energy of chemical bonds,
to thermal energy, and to energy for performing work. We know consider in general terms the transfer
of chemical bond energy.
The chemical reaction ADP + H
3
PO
4
ATP is associated with the generation of ATP energy.
The chemical energy of ATP phosphate bonds can be spent on osmotic, electric, mechanical, and other
types of work. In doing so, not all of the ATP energy is used for performing work; a portion of it is
dissipated as heat. All chemical processes in the living organism can proceed only with the involve-
ment of enzymes.

Bioenergetics
The energy resources stored in the cells are used to provide for the cells energetic requirements.
They include monosaccharides, amino acids, glycerol, and fatty acids. These materials, when crossing
the cytoplasmic membrane, may either be used immediately as energy sources or, by forming part of
biopolymers (polysaccharides, lipids, and proteins), be reserved as consumed for generating energy. In
the organism, the role of various tissues and organs involved in storing the energy sources, especially
such valuable ones as fats and carbohydrates, is different.

Stages of energy release from nutrients
In the process of energy release from various substrates, three conventional stages may be defi-
ned. The first stageis a preliminary one. This stage is necessary for converting biopolymers (supplied
in food or found in the cell) to a monomeric form suitable for energy extraction. This is accomplished
by hydrolyses in the intestine or within the cell. The intracellular hydrolysis is effected with the invol-
43
vement of cytoplasmic enzymes and lysosomes. The energetic value of this stage is rather inferior, sin-
ce only about 1% of the substrate energy is released, and it is dissipated as heat.
The second stageis partial degradation of monomers to key intermediates, chiefly to acetyl-ScoA
and to a number of the Krebs cycle acids (oxaloacetate and 2-oxoglutarate, or o-ketoglutarate). At the
second stage, a large number of initial substrates become reduced to three only. This stage is characte-
rized by partial (to 20%) release, under anaerobic (oxygen-free) conditions, of the energy contained in
the initial substrates. Part of this energy accumulates in ATP phosphate bonds; the other part is dissi-
pated as heat. The conversion of monomers proceeds in the hyaloplasm, and the final reactions reach
completion in the mitochondria.
The third stagerepresents an ultimate, oxygen-assisted degradation of the materials to CO
2
and
H
2
O. This phase comprises the aerobic biological oxidation of materials and proceeds with a complete
release of energy. The specific feature of chemical transformation at this stage is that out of the three
metabolites of the foregoing stage, after the so-called the Krebs cycle, only the hydrogen bound to car-
ries (NAD or FAD) remains. Hydrogen is the universal energy fuel which is employed in the respira-
tory chain for producing ATP and water. About 80% of the total chemical bond energy of compounds
involved is released at this stage. This energy, generated by the oxidation of substrates, is localized in
the ATP phosphate bonds, and a part of it is dissipated as heat. All the reactions of this stage are confi-
ned within the mitochondrial space.

Biological oxidation
Biological oxidation reactions are catalyzed by enzymes. The oxidation may be associated with:
- cleavage of hydrogen from the substrate to be oxidized (dehydrogenation);
- loss of an electron;
- addition of oxygen.
All of the three reaction types are equivalent and occur in the living cell.
Oxidation is not an isolated process, it is coupled to a reduction reaction, i.e. addition of hydrogen
or electron. Both compounds involved, i.e. the oxidant and the reductant, form a reduction-oxidation
pair, or redox-pair. The oxidative or reductive ability of a compound is characterized by its electron
affinity. The easier the substrate donates electrons, the higher is its reductive ability. On the contrary, a
high electron affinity of the substrate attests to its oxidative ability. The aptitude of a redox-pair for
reduction reactions is characterized by the standard oxidation-reduction potential, or redox potential.
This potential is expressed by the electromotive force (in volt, v) that develops in a half-element in
which the oxidant and reductant both have a concentration of 1,0 mol/liter at 25
o
C, pH=7,0 and are in
equilibrium with an electrode which can reversibly accept electrons from the reductant. The standard
redox potential of oxidation-reduction pair according to the reaction H
2
2H
+
+ 2e has been set at
zero. The redox potential for the system H
2
/2H
+
+ 2e is equal to 0,42v. The potential for the pair
NADH
+
+ H
+
/NAD
+
is 0,32v, which attests to a high ability of the pair to donate electrons, while the
redox potential for the pair O
2
/H
2
O shows a large positive value +0,81v; therefore, oxygen exhibits
the highest ability for accepting electrons. The redox-potential value enables one to predict a route for
electron transfer under biological oxidation conditions and to estimate the energy change on electron
transfer from a redox pair to another one.
As has been pointed out above, oxidation substrates are formed during the catabolic conversion of
proteins, carbohydrates, and lipids. These substrates are liable to dehydrogenation, the most common
type of biological oxidation with the involvement of intracellular dehydrogenases. The dehydrogena-
tion reactions that proceed with the participation of a substrate other than oxygen as a hydrogen accep-
tor, is referred to as the anaerobic oxidation. The biological oxidation reactions involving oxygen as a
hydrogen acceptor with the resultant production of water are known as tissue respiration.
The anaerobic oxidation is nothing other than hydrogen generation. Such reactions proceed with
the involvement of nicotinamide-dependent dehydrogenases (with NAD
+
and NADP
+
as acceptors for
the hydrogen split from an organic substrate), and flavin-dependent gehydrogenases (with FMN and
FAD as hydrogen acceptors). FAD can be reduced to FADH
2
, whereby it accepts two hydrogen atoms
(a net gain of two electrons):

44


Dehydrogenation substrates are formed in the extra-mitochondrial environment, but then are tran-
sported into the mitochondria within which oxidative metabolic processes take place.

Aerobic generation of energy in mitochondria
The major part of cellular energy is generated in the mitochondria, and for this reason these are
figuratively referred to as power plants of the cell. Mitochondria are organelles present in all plant and
animal cells. Mitochondria vary in shape and size, and outwardly look like an oval, rod, or thread. The
average size of mitochondria is about 0,53 m.
Any mitochondrion consists of two concentric membranes (membrane sacs). The inter-membrane
space filled with an aqueous medium lies between the outer and inner membranes.
The outer membrane is made up in half of proteins and lipids. In the inner membrane, proteins
account for about , and lipids, mostly cardiolipin, for about . The inner membrane is folded in a
characteristic manner to form cristae which extend into the mitochondrion inferior. The space between
the cristae is filled with an aqueous phase called matrix. On the matrix side of the inner membrane sur-
face there are attached small knob-like particles, earlier referred to as oxysomes.




Figure 22. Fifure of mitochondrium

Structure and function of respiratory chain
The hydrogen produced by dehydrogenation of substrates in the mitochondria becomes bound to
dehydrogenase-specific coenzyme carriers. The total of intra-mitochondrial hydrogen is supplied to
the respiratory chain via three channels. For the most part, hydrogen is bound with NAD as NADH
2

complexes; since the majority of intra-mitochondrial dehydrogenases are NAD-dependent. The next
part of intra-mitochondrial hydrogen is bound with NADP (as NADPH
+
+H
+
). The hydrogen supplied
through the third channel is produced by the action of flavin dehydrogenases (flavoproteins) on the
substrates, with FAD acting as a coenzyme.
The functional involvement of hydrogen bound to various coenzymes is different: NADH
2
and
FADH
2
are employed in oxidative processes, while NADPH
2
is chiefly used for reductive syntheses.
The inner mitochondrial membrane contains an enzyme, transhydrogenase, which controls the
distribution of hydrogen between NADP
+
and NAD
+
. Transhydrogenase catalyzes the hydrogen
transfer between the two nicotinamide-adenine-nucleotides:

NADPH
2
+ NAD
+
NADP
+
+ NADH
2


Tissue respiration and oxidative phosphorylation


45

Figure 23. Scheme of Respiratory chain

Hydrogen is utilized as the major fuel for energy generation. In the mitochondria, the flow of
electrons from hydrogen is channeled towards their terminal acceptor, oxygen. This results in the for-
mation of water which is the least efficient on the energy scale of biological materials and is the end
product of tissue respiration. It follows therefore that the tissue respiration is a redox process associa-
ted with formation of water due to the transfer of electrons from hydrogen onto oxygen.
The respiratory chain may be likened to a specific conveyer for the transport of protons and
electrons from the reduced NAD (NADH
+
+H
+
), which is formed by the action of NAD-dependent de-
hydrogenase on a substrate, or from the reduced FAD (FADH
2
), formed by action of flavin-dependent
dehydrogenases on a substrate, to oxygen.
The respiratory chain is made up of the following proton and electron carriers:
- flavoprotein-1 (FP) containing FMN for a coenzyme;
- coenzyme Q (ubiquinone);
- two iron-sulphur proteins containing nonheme iron (Fe);
- cytochromes b, c
1
, c, a, and a
3
.
In the liver, there occur about 5,000 respiratory chains per mitochondrion, and in the heart, about
20,000.

Complex I or NADH-Q oxidoreductase Complex II: Succinate-Q oxidoreductase

Flavoprotein (FP) is a NADH-dehydrogenase. This conjugated protein contains FMN acting as
an acceptor for protons and electrons supplied by NADH. Bound to flavoprotein is an iron-sulphur
protein likewise participating in the transport of protons and electrons onto CoQ. The active center of
NADH
+
-dehydrogenase is located on the inner side of the inner membrane; for this reason, dehydroge-
nation of NADH
+
proceeds at this particular side.
46

Figure 24. Reduction of coenzyme Q from its ubiquinone form (Q) to the reduced
ubiquinol form (QH
2
)

Coenzyme Q , or ubiquinone, is dissolved in the lipid layer of the membrane and can diffuse both
across and along the membrane.
Cytochromes b,c, c
1
, a, and a
3
,, are heme proteins. Cytochromes a and a
3
form a complex called
cytochrome oxidase. As distinct from other cytochromes, the cytochrome oxidase contains Cu
+.

As a matter of fact, tissue respiration in a simplified form resembles an explosive combustion of
hydrogen in oxygen according to the reaction:

2H
+
+ 2e + O
2
H
2
O

A coupling of respiration with phosphorylation is called oxidative phosphorylation. The P/O
ratio ia a measure for respiration-phosphorylation coupling. This ratio was called phosphorylation
ratio. The uptake of one oxygen atom (or transfer of an electron pair from a substrate to oxygen) pro-
ceeded with the involvement of about three, rather than one, inorganic phosphate moieties, i.e. the P/O
or P/2e, ratio was equal to about 3.

Complex III: Q-cytochrome c oxidoreductase
Other vise stated, the respiratory chain possessed a minimum of three coupling, or phosphoryla-
tion, sites involved in the formation of ATP with the participation of inorganic phosphate according to
the scheme:
ADP + H
3
PO
4
ATP + H
2
O


47

Complex IV: cytochrome c oxidase H
+
- ATP synthase

For the formation of one macroergic ATP bond with energy expenditure of at least 40 Kj/mol, a
redox-potential drop of about 0,22 v per electron pair transferred between the respiratory chain steps is
required: E= 0,22 v.
Agents affecting energy metabolism in cells
Numerous materials, including toxins and therapeutic drugs, are capable of affecting the cell ener-
getic through intervening in the energy-generating glycolysis and oxidative phosphorylation processes.
Isonicotinic acid hydrazides (phthivazid) are competitive inhibitors for NAD-dependent dehydro-
genases. Malonate is a competitive specific inhibitor for succinate dehydrogenase.



Rotenone
Amytal, rotenone (fish poison), and progesterone (female sex hormone) inhibit the first coupling
link (proton potential generation) and render inoperative the portion of respiratory chain ahead of the
block.
Cyanides (NaCN, KCN), azides (NaN
3
), and carbon monoxide (CO) inhibit cytochrome oxidase
and render thereby the very process of respiration impossible.

Respiration-phosphorylation coupling mechanism in mitochondria
A basically novel mechanism for respiration-phosphorylation coupling has been developed by the
British biochemist Mitchell (1961). The hypothesis that has been put forward by him is currently kno-
wn as chemiosmotic, or proton-driving, hypothesis. According to Mitchell, the energy of electron/pro-
ton transfer along the respiratory chain is initially stored as a proton potential, or H
+
ion electrochemi-
cal gradient which is generated as the protons move across the membrane. The reverse proton diffusi-
on across the membrane is coupled to phosphorylation, which is accomplished by H
+
-ATP-synthetase.
Respiration performs an osmotic work (i.e. makes the protons concentrate in the extramitochondrial
medium) and an electric work (by generating an electric potential difference), both of which are con-
verted by ATP-synthetase into a chemical stored energy, i.e. the synthesis of ATP takes place. The
junction of these two functions, respiration and phosphorylation, has provided a good reason to name
the hypothesis chemiosmotic, or proton-driving, since here the proton potential is a driving force for
phosphorylation.

LECTURE 10

Subject: ENZYMIC MITOCHONDRIAL SYSTEMS AS HYDROGEN
GENERATORS

Pyruvate, the anion of pyruvic acid, is an important oxidation substrate formed as a metabolic in-
termediate from carbohydrates, proteins, amino acids, and glycerol. Pyruvate is subjected to oxidation
in the mitochondria where it penetrates from the cytoplasm. In addition to pyruvate, a number of subs-
trates undergo mitochondrial oxidation. Some of them are involved in accepting cytoplasmic hydrogen
and transferring it within the mitochondria to the respiratory chain. The significance of pyruvate as an
oxidation substrate resides also in that it can be oxidatively decarboxylated to acetyl-ScoA, a major
producer of hydrogen in the mitochondria. For that matter let us consider the enzymic system of pyru-
vate oxidation.

Oxidation of pyruvate to acetyl-ScoA
48
The oxidative decarboxylation of pyruvic acid is effected by a multienzyme pyruvate dehydroge-
nase complex. This complex is found in the matrix not in a dissolved state, but rather attached to surfa-
ce proteins of the inner mitochondrial membrane exposed to the matrix. The pyruvate dehydrogenase
complex exemplifies the structural organization of a number of different enzymes, and features all the
advantages of such an organization. The complex is composed of three enzymes: pyruvate dehydroge-
nase (E
1
), dihydroxylipoylacetyl transferase (E
2
), and dihydrolipoyl dehydrogenase (E
3
). Pyruvate de-
hydrogenase is made up of 24 enzyme molecules, each containing a thiamine diphosphate (TDP or
TPP) residue acting as coenzyme for pyruvate dehydrogenase. The quaternary structure of dihydroli-
poylacetyl transferase includes 24 subunits. Each subunit of dihydrolipoylacetyl transferase contains a
lipoic acid (LA) residue:

The complex includes 12 molecules of dixydroxylipoyl dehydrogenase, each containing a FAD
residue. In addition, during the oxidation of pyruvate, two outer (complex-nonbound) coenzymes,
coA-SH and NAD, are involved, which act as acceptors for the pyruvate oxidation products.


Coenzyme A
The pyruvate is successively acted on by the enzymes of the pyruvate dehydrogenase complex.
The following stages for pyruvate oxidation may be considered:

Figure 25. PDHcomplex reactions

The overall reaction for oxidation of pyruvate with the pyruvate dehydrogenase complex enzy-
mes is expressed as:




Practically, the total of the pyruvate supplied to the mitochondria is rapidly oxidized to acetyl-
ScoA. Of the pyruvate oxidation products, the end metabolite CO
2
is of little energetic value. In con-
trast, the reduced NAD is an energy-rich material which supplies hydrogen to the respiratory chain
(NADH
2
=3ATP). Acetyl-ScoA is engaged in the Krebs cycle operative in the mitochondria.
Oxidative enzyme system of the Krebs cycle
49
Krebs cycle is a major enzymic system acting as a hydrogen generator for the respiratory chain.
In 1937, Krebs, a German biochemist in England suggested, on the basis of his experiments and the
data obtained by Szent-Guorgyi, that an oxidative cyclic system of reactions might be operable in the
cells. He proposed the name citric acid cyclebecause at that time there was no reliable evidence that
citric acid could be the first substrate of the cycle. As has been shown later, this cycle is the major en-
zymic system for oxidation of citric acid residue (acetyl-coA) and that the first reaction of the cycle is
the synthesis of citric acid. However, most commonly this cycle is referred to as the Krebs as a tribute
to Sir Hans Adolf Krebs who was the first to establish the reaction sequence in this cycle.
Separate reactions involved in the Krebs cycle
Acetyl-ScoA, produced by oxidation of pyruvate, fatty acids, and amino acids, is included in the Krebs
cycle.
Biochemical functions of the Krebs cycle
Integrative function: the Krebs cycle acts as a specific metabolic collector that unifies the cata-
bolic pathways of carbohydrates, lipids, and proteins.
Amphibolic function: the Krebs cycle performs a dual function: catabolic one, since the cycle sub-
strates are used in the synthesis of other materials. For example, oxaloacetate is utilized in the synthe-
sis of glutamic acid; and succinate, in the synthesis of heme.
Energetic function: in the course of the Krebs cycle reactions, one substrate level ATP molecule
is formed per 1 mole of acetyl-ScoA supplied.

























Figure 26. Reactions of the Krebs cycle

Hydrogen-donating (or hydrogen-generating) function: the Krebs cycle is a major hydrogen ge-
nerator for the respiratory chain. In the Krebs cycle, four pairs of hydrogen atoms are formed, of whi-
ch three pairs are bound with NAD (3NADH
+
=9ATP), and one, with FAD (FADH
2
=2ATP).
The terminal stage of the Krebs cycle merits a closer look. Firstly, it should be kept in mind that
all the processes that supply the Krebs cycle with acetic acid residues (acetyl-ScoA) and other inter-
mediates (di- and tricarboxylic acids), ensure the operation of the Krebs cycle and its function of gene-
rating hydrogen for the respiratory chain. These processes include the cycle for oxidation of fatty acids
and pyruvate (as sources of acetyl-ScoA) and the reactions for degradation of the amino acid carbon
skeleton (as sources of acetyl-ScoA and dicarboxylic acids).
50

LECTURE 11

Subject: CARBOHYDRATES: CHEMICAL STRUCTURE, DIGESTIVE
MECHANISM OF SUGARS

Carbohydrates are the most widespread compounds involved in the buildup and biological func-
tions of the cell. Carbohydrates (sugars) are polyhydroxycarbonyl compounds and their derivatives.
The term carbohydrate is rather obsolete and reflects properly neither the chemical nature nor the
composition of the species in question; still, the suggested alternative term glycide has not gained
acceptance. A characteristic feature of carbohydrates is the occurrence in them of a carbonyl (aldo or
keto) group and at least two hydroxyl groups. Therefore, glyceraldehyde and dihydroxyacetone may
be referred to as the simplest carbohydrates:

H-C=O CH
2
-OH
| |
H-C-OH C=O
| |
CH
2
-OH CH
2
-OH

glyceraldehyde dihydroxyacetone

The classification of carbohydrates is based on their structure and physico-chemical properties.
Carbohydrates bearing an aldo group are called aldoses and those bearing a keto group, ketoses.
Based on their physico-chemical properties, carbohydrates are divided into:
- neutral species (those containing hydroxyl and carbonyl groups only: glucose),
- basic (those containing, alongside the aforementioned groups, an amino group: aminosacchari-
des),
- acidic (those containing, except hydroxyl and carbonyl groups, carboxyl moieties also: glucu-
ronic acid).
Based on their chemical structure, carbohydrates are divided into:
- monosaccharides, or monoses, which are simple carbohydrates,
- olygosaccharides, which are carbohydrates possessing two to ten monosacchride units linked
by glycoside bonds,
- polysaccharides, or glycans, which are high-molecular carbohydrates containing more than ten
monosaccharide units linked by glycoside bonds.
Monosaccharides, or monoses, are simple carbohydrates. The names of all monosaccharides end
in ose. The group name of monosaccharides indicates the number of carbon atoms and the presence
of an appropriate carbonyl group (aldo or keto). For example, monosaccharides possessing five carbon
atoms are called pentoses and taking into account the occurrence of an aldehyde group, aldopentoses.
If the monosaccharides in question contain a keto group, they are called ketopentoses. Carbohydrates
with six carbon atoms are called, respectively, aldohexoses and ketohexoses.
Monosaccharides belong to either L-(levorotatory) or D-(dexterorotatory) series depending on the
L- or D-configuration of substituents at the asymmetric carbon atom farthest from the carbonyl group
in a given monosaccharide. For example, in hexoses, this carbon is C-5.
In aqueous solution, monosaccharides exist either in the form of an unfolded chain, or as a cyclic
structure. Cyclic monosaccharides do not occur in trioses and tetroses but, starting with pentoses, a
spontaneous reaction takes place between one of the hydroxyl groups and the carbonyl group leading



51
D-Glucose (aldohexose) D,o-Glucose depicted in Haworth projection


Figure 27. Chemical structures of carbohydrates-monosaccharides
to the closure of a ring. In such a manner, for example, five-membered (furanose) and six-membered
(pyranose) rings are formed. The hydroxyl at the first carbon atom of a cyclic monosaccharide is cal-
led the half-acetal hydroxyl. It is essential for reducing properties of carbohydrates.
Monosaccharide derivatives. The modification of available functional groups, or the introduc-
tion of substituents into the molecule of a monosaccharide produces various monosacchride derivati-
ves. These are used in generating diverse polymeric carbohydrates. Some of the derivable species may
act as intermediary metabolites.
2-deoxy-D-ribose (or simply: deoxyribose) forms part of deoxyribonucleosides and deoxyribonu-
cleotides, which are structural monomers of DNA.
Many monosaccharides are involved in the synthesis of a very important group of compounds
called glycosides. In particular, ribose and deoxyribose make part of the aforementioned nucleosides
and nucleotides which are N-glycosides. Of importance is the role of aminosugars, or aminodeoxysu-
gars. D-glucosamine is used in the buildup of essential structural polysaccharides, i.e. hyaluronic acid.
D-galactosamine takes part in the synthesis of polysaccharides of cartilage tissue, chondroitin sulpha-
tes and certain glycolipids.

Biological importance of monosaccharides
In the cell, the monosaccharides are used as a source of energy. Monosaccharides (as well as po-
lysaccharides) as distinct from other compounds, are an energy substrate for the cells of human and
animal organisms both in the presence and in the absence of oxygen. Moreover, monosaccharides and
their derivatives are involved in the buildup of diverse biological molecules, i.e. in the performance of
a plastic function.



52
Figure 28. Chemical structure of monosaccharide derivatives

Olygosaccharides, are carbohydrates possessing two to ten monosaccharide units linked by gly-
coside bonds. They differ from one another in the monosaccharide composition and in the type of gly-
coside bond. Among the most widespread olygosaccharides worth mentioning are:
- sucrose (cane sugar, beet sugar) widely distributed in plants,
- maltose (malt sugar), a product of partial hydrolysis of starch in plants and glycogen in
animals,
- lactose (milk sugar) found in the milk of all mammals,
- trehalose, present in numerous lower and higher fungi.



Figure 29. Chemical structures of disaccharides: sucrose (left) and lactose

Olygosaccharides are found in the cells and biological fluids in a free state as well as constituents
of covalently bonded carbohydrate-protein complexes (glycoproteins and proteoglycans).
Polysaccharides, or glycans, are high-molecular carbohydrates containing more than ten monosa-
ccharide units linked by glycoside bonds. Polysaccharides differ from one another in the nature of mo-
nosaccharides involved, in molecular mass, and in the type of chain-linking glycoside bonds. The most
common monomeric unit in polysaccharides is D-glucose. Other monosaccharides that occur are
galactose, mannose, fructose, and monosaccharide derivatives.
Distinguished are homopolysaccharides (homoglycans) built of only one type of monosacchari-
des and heteropolysaccharides (heteroglycans) composed of different types of monosaccharides. For
example, starch is a homopolysaccharide, since it contains D-glucose only, while hyaluronic acid is a
heteropolysaccharide, since it is made up of alternating units of D-glucuronic acid and N-acetyl-D-glu-
cosamine. Polysaccharides are also subdivided into linear and branched species, according to the
structural type of their polymeric chains.

General properties of polysaccharides
Polysaccharides exhibit typical properties common to high-molecular compounds bearing polar
groups. Thence, polysaccharides are hydrophilic; when placed in water, they swell (similar to fibrous
proteins) and then dissolve partially to form colloidal solutions.
Polysaccharides are contained both inside the cells and in the extracellular matter. This signifies
that the characteristic properties of polysaccharides show up under a variety of conditions. As a rule,
neutral polysaccharides inside the cells are intended as reserve material (starch or glycogen). Acidic
polysaccharides (hyaluronic acid and chondroitin sulphate) are commonly found in the extracellular
space.

Individual polysaccharides
Neutral polysaccharides. The structures for the most widespread homopolysaccharides are pre-
sented below:
53

Figure 30.Glycogen structure

Starch is a homopolysaccharide found in plants; it consists of o-amilose and amylopectin. Glyco-
gen is the major short-term storage polysaccharide found in all human and animal tissues. In small
amounts, glycogen is also present in bacteria and plants. Cellulose is a structural homopolysaccharide
of plants.
Acidic heteropolysaccharides, or mucopolysaccharides (from the Latin mucus, slime). They owe
this name to their slime-like consistency. These polysaccharides are highly hydrated, gel-like sticky
materials bearing a distinct negative charge. They all are found in the extracellular material, not in a
free state but rather bound with proteins. Such heteromacromolecules are referred to as proteoglycans
or glucosamine proteoglycans, since the basic properties of these macromolecules are mainly due to
the carbohydrate, rather than the protein, moiety (to be noted, the functional behavior of glycogen-pro-
tein and starch-protein complexes is also determined by the carbohydrate component).
The acidic heteropolysaccharides, depending on their chain structure, are divided into seven ma-
jor types. Six of them, represented by hyaluronic acid, chondroitin 4-sulphate, chondroitin 6-sulphate,
dermatan sulphate, heparin, and heparan sulphate, possess similar repeating disaccharide units (for
example: D-glucuronic acid, N-acetyl-D-glucosamine, D-galactose, etc.).
Hyaluronic acid is a nonsulphated linear heteropolysaccharide. It has the greatest molecular mass
among the heteropolysaccharides. It serves as the biological cement filling the intercellular space. The
molecules of hyaluronic acid a gel-like network which acts as a biological filter retaining microbiotic
and other large molecules that invade the organism.




















Figure 31. Glucosoamineglycans

Chondroitin sulphates are the most abundant acidic heteropolysaccharides in human and animal
tissues. They occur in skin, bone tissue, cartilage, in tissues of trachea, aorta, arteries, etc. In tissues,
54
they are bound to the protein basis, or protein core, which makes up about 17-22% of the heteromole-
cule mass.
Dermatan sulphate is contained in the aorta and in distinction from other chondroitin sulphates,
possesses anticoagulative properties.
Keratan sulphate is found in the cornea, where it is bound covalently to protein. Heparin and
heparan sulphate are structurally related species. As distinct from other acidic heteropolysaccharides,
they are not structural components for the extracellular material. In blood heparin is noncovalently
bound to specific proteins. The complex of heparin with glycoprotein of plasma exhibits an anticoagu-
latory activity, while the complex of heparin with the enzyme lipoprotein lipase is capable of cleaving
lipids found in the blood as chylomicrons.
Biological functions of polysaccharides
The essential functions of polysaccharides are:
- energetic
- supportive
- protective
- colligative (structural)
- hydroosmotic and ion-regulating
- cofactor.
Digestive mechanism for carbohydrates
The digestion of carbohydrates starts in the oral cavity, mainly through the agency of salivary o-
amylase. Certain dietitians believe that another enzyme, maltase, is also present in the saliva. o-Amy-
lase is composed of a single polypeptide chain; it is stabilized by calcium and activated by chloride
ions, its optimum pH is 7,1. The enzyme belongs to endoamylases and acts on the endo o-1,4-glycosi-
de bonds of dietary starch and glycogen. It is incapable of hydrolyzing the o-1,6-glycoside bonds of
these polysaccharides. By the action of salivary o-amylase, the polysaccharides are split into a limit-
dextrin, maltose, and a small amount of glucose. Dietary disaccharides, the major ones of which are
sucrose, lactose, suffer no cleavage in the oral cavity.
In the stomach, o-amylase is inactivated by gastric acid components, and digestion of carbohydrates
ceases. Hydrolysis of polysaccharides, including o-limitdextrin, formed in the oral cavity, and of
disaccharides to monosaccharides proceed in intestine. The enzymic activity is favoured by neutrali-
zation of acidic food by hydrocarbonates dissolved in the alkaline contents of pancreatic juice and bile.
The intestinal hydrolysis of carbohydrates is carried out by the enzymes of pancreas and intestine.
The former enzymes are pancreatic o-amylase and oligo-1,6-glucosidase. The hydrolytic action of
pancreatic o-amylase is similar to that of salivary o-amylase. Within a short span of time (4-5 minu-
tes), pancreatic o-amylase hydrolyzes the delivered starch and glycogen to o-limit-dextrin and malto-
se. Hydrolysis of o-limit-dextrin is effected through the aid of oligo-1,6-glucosidase, which is capable
of specifically disrupting o-1,6-glycoside bonds at branch points of polysaccharide. Maltose is for-
med as the end product. Disaccharides undergo hydrolysis in the intestinal wall, rather than in the in-
testinal cavity, and the monosaccharides formed are immediatelly absorbed.
o-Oligosaccharidases include maltase, sucrase, lactase:
Maltase
Maltose + H
2
O 2 glucose
Sucrase
Sucrose + H
2
O glucose + fructose
Lactase
Lactose + H
2
O glucose + galactose
The end products of carbohydrate digestion are monosaccharides, mostly glucose, fructose, and
galactose. Further absorption of monosaccharides occurs in the small intestine.
The uptake of monosaccharides produced by digestion of carbohydrates is accomplished via the
secondary active transport (K, Na-ATPase).


55

Na
+
/K
+
ATPase

The transport of monosaccharides is dependent on Na
+
ions and is mediated by a special carrier,
the Na
+
gradient being the driving force for the monosaccharide transport, like in the amino acid ab-
sorption. The absorption rates for separate monosaccharides (hexoses and pentoses) differ. Galactose
is absorbed at the highest rate, followed by glucose. The absorbed monosaccharides are delivered from
the intestinal wall into the portal vein, to the liver, and then are supplied in the blood to other tissues.
In the liver, the rest of hexoses (galactose, fructose and mannose) are converted to glucose or its meta-
bolites. In addition to the liver, the major consumers of glucose are the brain and skeletal muscles
wherein glucose is utilized as an easily oxidizable source of energy. In the adipose tissue, glucose is
employed for the synthesis of neutral fat. Usually, about 65% of glucose supplied from the intestine is
consumed for oxidation in the cells (for generation of energy). Further 30% and 5% are used, respecti-
vely, in the syntheses of fat and glycogen. These percentages may vary, depending on the physiologi-
cal state of the organism, age, and a number of other factors.

LECTURE 12

Subject: GLYCOLYSISANAEROBIC OXIDATION OF CARBOHYDRATES

Energy in the cells is produced not only by oxidative phosphorylation (via the aerobic route), but
also by degradation of nutrients, omitting the intervention of molecular oxygen. Such an oxygen-free,
or anaerobic, route to energy generation is referred to as fermentation. Fermentation is a pristine and
simplest mode of energy extraction as compared with the aerobic route. In the cells hexoses are the
major source of energy generation by the anaerobic route. Here, of prime importance is D-glucose.

Degradation of glucose: glycolysis
In the cells of humans and other higher organisms (animals and plants) lactic acid fermentation,
commonly referred to as glycolysis (from the Greek glykis, sweet, and lysis, degradation, decom-
position) takes place. Thus, glycolysis is the anaerobic degradation of glucose to two molecules of lac-
tic acid.
Separate steps of glycolysis are catalyzed by 11 enzymes. These form a chain of functionally in-
terrelated enzymes (multienzyme functional system), in which the product of a reaction catalyzed by
an enzyme serves as a substrate for the enzyme at the next step. Glycolytic enzymes are found outside
the mitochondria, in the cell sap, where they either persist in a dissolved state, or are loosely bound to
the endoplasmic reticulum membrane. Degradation of glucose is an exergonic process. The energy
liberated in this process accumulates in the ATP phosphate bonds.

Separate reactions of glycolysis
1. Phosphorylation of D-glucose. This is very starting reaction that triggers the glycolysis.
2. Isomerization of glucose-6-phosphate to fructose-6-phosphate.
3. Phosphorylation of fructose-6-phosphate to fructose-1,6-biphosphate.
Phosphofructokinase is a key enzyme in the glycolysis, since, being the ratelimiting factor for
the overrall process (due to reaction irreversibility), is controlled by various iso- and allosteric regula-
tors.
56
4. Cleavage of fructose-1,6-biphosphate into glyceraldehyde-3-phosphate and dihydroxyacetone
phosphate.
Fructose-1,6-biphosphate splits into two phosphotrioses; because of this, glycolysis was earlier
referred to as the dichotomic route of glucose conversion.
5. Interconversion of triose phosphates.
6. Oxidation of glyceraldehyde-3-phosphate to 1,3-biphodphoglycerate.
7. Transfer of phosphate group from 1,3-biphosphoglycerate onto ADP (first glycolytic phospho-
rylation).
8. Isomerization of 3-phosphoglycerate to 2-phosphoglycerate.
9. Dehydration of 2-phosphoglycerate to phosphoenolpyruvate.
10. Transfer of phosphate group from phosphoenolpyruvate onto ADP (second glycolytic phos-
phorylation).
11. Reduction of pyruvate to lactate.
This reaction is catalyzed by concluding enzyme of glycolysis, lactate dehydrogenase (for short
LDH).
In human and animal tissues, five lactate dehydrogenase isoenzymes occur whose composition is
defined by the specific features of oxidative tissue metabolism. Isoenzymes LDH
1
and LDH
2
(H type,
heart-muscle) are effective under aerobic conditions, when pyruvate undergoes a fast oxidation, rather
than a reduction to lactate. In contrast, isoenzymes LDH
4
and LDH
5
(muscle, of M type) are effective
under anaerobic conditions.
Glucose is transformed to lactate via the glycolytic process. Lactate is an impasse in the meta-
bolic pathway, since it is incapable of participating in any biochemical process, except the reverse
conversion to pyruvate. When lactate is accumulated in the cells, the intracellular medium pH may be
disturbed and the glycolysis suppressed. For this reason, lactate is scavenged from the cells as the
metabolic ballast. However, in certain organs (for example, heart) it is amenable to oxidation and is
used as an energetic material.
Energy balance and biological function of glycolysis
Glycolysis may be regarded as an internal redox process involving the formation of two molecu-
les of NADH
2
at the glyceraldehyde-3-phosphate dehydrogenation step with the subsequent acceptan-
ce of hydrogen by two pyruvate molecules to produce lactate. The overall scheme for glycolysis is
shown below:
Glucose + 2H
3
PO
4
+ 2ADP 2 lactate + 2ATP + H
2
O























57


















Figure 32. Glycolysis

At the steps of glucose and fructose phosphorylation (steps 1 and 3), two ATP molecules are con-
sumed. Taking into account that the total of dihydroxyacetone phosphate (at step 5) is converted to
glyceraldehyde-3-phosphate, i.e. the reaction yields a molecule of glyceraldehyde-3-phosphate more
to the available one, the further route is actually a successive conversion of the two molecules of this
phosphotriose. Therefore, at each of the two steps of glycolytic phosphorylation catalyzed by phos-
phoglycerate kinase and pyruvate kinase, two ATP molecules are formed. In total, the net yield in
ATP is two molecules per molecule of split glucose. The energetic value of glucose is thus readily per-
ceived.
In critical situations in the organism, when for some reasons the delivery or consumption of oxy-
gen is hindered, glycolysis is the only way to urgent energy supply for sustaining the vital activity of
the cells. Therefore, in hypoxia (reduction of oxygen supply to tissues), glycolysis plays an important
energetic role. Glycolysis is operable in all cells and tissues; commonly, its potentiality is superior to
the need in carbohydrate cleavage, i.e. glycolysis enzymes are present in the cells in excess. For the
erythrocytes that lack mitochondria, glycolysis remains the only possibility to produce ATP and main-
tain the functional and structural integrity.















Figure 33. Glycerophosphate shuttle mechanism

58

Figure 34. Malate-aspartate shuttle mechanism

The yield in energy under aerobic metabolism conditions amounts to 38 ATP molecules per
molecule of split glucose provided the cytoplasmic NADH
2
hydrogen is supplied to the respiratory
chain via malate-aspartate cycle (via glycerophosphate shuttle mechanism 36 ATP).

Degradation of glycogen (glycogenolysis)
In the mammalian cells, glycogen is a reserve carbohydrate whose breakdown produces energy.
The process by which glycogen is degraded is called glycogenolysis. The breakdown of glycogen is
accomplished via phosphorolysis and hydrolysis. Phosphorolysis of glycogen is carried out through
the aid of glycogen phosphorylases and oligo-1,6-glycosidase, an accessory enzyme in this process.
Mechanism for phosphorylation of phosphorylase B (according to Sutherland):



Figure 36. Adenylate cyclase mechanism













Biosynthesis of glycogen (glycogenogenesis)
Synthesis of glycogen is carried out in all cells of organism, but this process is especially active in
skeletal muscles and in the liver. Two routes for glycogen synthesis are possible. One route involves a
successive addition of glucose units to the extant glycogen moiety (glycogen primer); the other one
originates in glucose molecules. The source of glucose, uridine diphosphate glucose (UDP-glucose),
which is produced from glucose-1-phosphate and uridine triphosphate (UTP) through the agency of
the enzyme glucose-1-phosphate uridyltransferase according to the scheme:

Glucose-1-phosphate + UTP UDP-glucose + H
4
P
2
O
7









59
























Figure 35. Structure of glycogen
The next step involves a transfer of the glucose residue from UDP-glucose onto the glycogen
primer through the aid of the enzyme glycogen synthetase:

UDP-glucose + (Glucose)
n
UDP + (Glucose)
n+1








To be noted, the glycogen synthetase catalyzes the formation of o-14 glycoside bonds only.
The branching enzyme, amylo-(o-1,4o-1,6)-trans-glycosylase, transfers short fragments (two
or three glucose residues) from one portion of the glycogen molecule onto another and forms o-16-
glycosidic bonds (branch points). The alternating action of these two enzymes results in the lengthe-
ning of the glycogen molecule.

Gluconeogenesis
The synthesis of glucose from noncarbohydrate sources is referred to as the gluconeogenesis. It is
feasible only in certain organism tissues. The major site for gluconeogenesis is the liver.
Mechanism for gluconeogenesis. Since the glycolysis involved three energetically irreversible
steps at the pyruvate kinase, phosphofructokinase, and hexokinase levels, the production of glucose
from simple noncarbohydrate materials , for example, pyruvate or lactate, by a reversal of glycolysis
(from bottom upwards) is impossible. Therefore, indirect reaction routes are to be sought for:
1. The first indirect route in glucose synthesis involves the formation of phosphoenolpyruvate
from pyruvate without the intervention of pyruvate kinase. This route is catalyzed by two enzymes:

CH
3
+ HCO
3
+ ATP pyruvate carboxylase COOH
| |
C=O C=O + ADP + H
3
PO
4

| |
COOH CH
2

pyruvate |
COOH
oxaloacetate
60
COOH COOH
| + GTP (ATP) | + CO
2
+ GDP (ATP)
CH
2
phosphoenolpyruvate- C-O~PO
3
H
2

| carboxylase
C=O CH
2

|
COOH phosphoenolpyruvate
oxaloacetate

2. The second indirect route involved the formation of fructose-6-phosphate from fructose-1,6-
biphosphate without the intervention phosphofructokinase reaction. This route is catalyzed by fructose
biphosphatase:
fructose-1,6-biphosphate + H
2
O fructose-6-phosphate + H
3
PO
4

3. The third indirect route involves the formation of free glucose from glucose-6-phosphate by
circumventing the hexokinase reaction. This route is catalyzed by glucose-6-phosphatase:

glucose-6-phosphate + H
2
O glucose + H
2
O

The free glucose produced by this reaction is supplied to the blood from the tissues. Amino acids
involved in the gluconeogenesis are referred to as glycogenic amino acids (for example, alanine, seri-
ne, cysteine, etc.).

Alcoholic fermentation
This route of carbohydrate fermentation diverges from the glycolysis route at the step of pyruvate
conversion:
CH
3
pyruvate CH
3
alcohol CH
3

| decarboxilase | dehydrogenase |
C=O C=O CH
2

| -CO
2
| NADH
2
NAD |
COOH H OH

pyruvate acetaldehyde ethanol


CH
3
CH
3
CH
3

| alcohol dehydrogenase | aldehyde dehydrogenase |
C=O C=O C=O
| NAD NADH
2
| NADH
2
NAD |
OH H OH

ethanol acetaldehyde acetate

Alcoholic fermentation is typical of yeast and certain microorganisms. Alcohol dehydrogenase,
present in human tissues (especially in liver) oxidizes ethanol to acetalaldehyde; the latter becomes in-
volved in the metabolism through aldehyde dehydrogenase.
Acetate enters the Krebs cycle in an activated form:
Acetate + CoA-SH + ATP acetyl-coA synthetase acetyl ScoA +AMP +PP

LECTURE 13

Subject: PENTOSE PHOSPHATE CYCLE. CARBOHYDRATE
METABOLISM CONTROL IN THE HUMAN ORGANISM

There is known one more catabolic route for carbohydrates commonly referred to as the pentose
phosphate cycle(also called hexose monophosphate shunt, or phosphogluconate pathway). The pento-
se phosphate cycle represents a multienzyme system in which the important intermediates are, the na-
me implies, pentose phosphates. This cycle may be regarded as a branching, or shunt, at the glucose
6-phosphate step in the overall glycolysis. To provide for all steps of the pentose phosphate cycle, at
least three glucose-6-phosphate molecules are required.

61








Figure 37. Reactions of pentose phosphate cycle (PPC)

Then fructose-6-phosphate and glyceraldehyde-3phosphate produce and enter the glycolysis.
Thus, in the course of reactions catalyzed by the intrinsic enzymes of the pentose phosphate cycle, two
fructose-6-phosphate molecules, one glyceraldehyde-3-phosphate molecule, and three carbon dioxide
molecules are produced from three glucose-6-phosphate molecules. In addition, six NADPH
2
molecu-
les are formed. The overall scheme for the pentose phosphate cycle is:

3 Glucose-6-phosphate + 6NADP
+

2 Fructose-6-phosphate + glyceraldehyde-3-phosphate + 6NADPH
2
+ CO
2


Interrelation of the pentose phosphate cycle and glycolysis
These two pathways for carbohydrate conversion are closely related. The products of the pentose
phosphate route fructose-6-phosphate and glyceraldehyde-3-phosphate are likewise glycolysis
metabolites; for this reason, they are involved in glycolysis and undergo conversion by glycolytic
enzymes. Two molecules of fructose-6-phosphate can regenerate to two glucose-6-phosphate molecu-
les through the agency of the glycolytic enzyme glucose-phosphate isomerase. Here, the pentose phos-
phate pathway functions as a cycle. The other product, glyceraldehyde-3 phosphate, enters the glyco-
lysis to be either converted to lactate (under anaerobic conditions) or oxidized to CO
2
and H
2
O (under
aerobic conditions). As can easily be estimated, the conversion of glyceraldehyde-3-phosphate to lac-
tate leads to the formation of two ATP molecules, while the combustion to CO
2
and H
2
O produces 20
ATP molecules. It follows there for that under physiological conditions, when the pentose phosphate
pathway for carbohydrate conversion is included in the glycolysis, the overall process of glucose-6
phosphate conversion may be expressed via the pentose phosphate cycle.
Under anaerobic conditions:

3 glucose-6-phosphate + 6NADP
+
+ 2H
3
PO
4

2 glucose-6-phosphate + lactate + 2 ATP + 6NADPH
2
+ 3CO
2


Under aerobic conditions:

3 glucose-6-phosphate + 6NADP
+
+ 20ADP + 20H
3
PO
4

2 glucose-6-phosphate + 6NADPH
2
+ 6CO
2
+ 6H
2
O + 20ATP

At the first glance, the energetic value of this conversion of glucose-6-phosphate via the pentose
phosphate cycle appears to be inferior to that of the aerobic glycolysis pathway, the latter providing a
maximum of 38 ATP molecules. However, it should be borne in mind that a major portion of energy is
stored in NADPH
2
, and 6 NADPH
2
molecules are energetically equivalent to 18 ATP molecules. Con-
sequently, the energetic effect remains the same.

The biological function of the pentose phosphate cycle
The biological function of the pentose phosphate cycle involves the production of two com-
pounds: NADPH
2
, which is a reductive force in the synthesis of various materials, and the metaboli-
te ribose-5-phosphate, which is used as a building material in the synthesis of various species. The ma-
jor functions of the pentose phosphate cycle are:
- amphibolic function: the cycle is a route to degradation of carbohydrates and, simultaneously,
to the supply of materials used in synthetic reactions (NADPH
2
and ribose-5-phosphate);
- energetic function, since the involvement of pentose phosphate cycle products (glyceraldehy-
de-3-phosphate) in the glycolysis produces energy;
- synthetic function, as a major function associated with the use of NADPH
2
and ribose-5-phos-
phate.
62
NADPH
2
is used:
- in the detoxification of drugs and poisons in the monooxygenase oxidation chain of the endo-
plasmic reticulum of the liver;
- in the synthesis of fatty acids and other structural and reserve lipids;
- in the synthesis of cholesterol and its derivatives bile acids, sterol hormones (corticosteroids,
female and male sex hormones), and vitamins D;
- in the neutralization of ammonia under reductive amination.
Ribose-5-phosphate is used in the synthesis of histidine, nucleosides and nucleotides (nucleotide
mono-, di-, and triphosphates), nucleotide coenzymes (NAD, NADP, FAD, and HS-coA), and poly-
meric nucleotide derivatives (DNA, RNA, and short-chain oligonucleotides). A high activity of this
pathway is observed in fat tissue, liver mammary gland (especially during lactation), adrenal glands,
gonad glands, marrow, and lymphoid tissue. Relatively high is the activity of pentose phosphate shunt
dehydrogenases in the erythrocytes. A low activity of the pentose phosphate pathway is observed in
muscular tissue (heart and skeletal muscle).

Carbohydrate metabolism control in the organism
From the standpoint of activity of the whole organism, certain specializations of the carbohydrate
metabolic routes in individual tissues are profitably complementary. For example, strenuous muscular
exertion requires energy which is initially supplied by the breakdown of glycogen to lactic acid. The
latter compound is excreted into the blood to be supplied to the hepatic tissue, where it is used for the
synthesis of glucose during gluconeogenesis. From the liver, glucose is delivered in the blood to the
skeletal muscles to be consumed for energy generation and to be deposited as glycogen. This intertis-
sue (or interorgan) cycle in the carbohydrate metabolism is referred to as the Cori cycle (called also
glucose-lactate cycle).


Figure 38. Cori cycle

The maintenance of a constant glucose level in the blood is of primary importance for the orga-
nism, since glucose is the major energy substrate for the nervous tissue. The normal glucose content in
the blood is 3,3 to 5,5 mmol/liter.
An increased concentration of glucose in blood is known as hyperglycemia. If hyperglycemia rea-
ches as high as 9 to 10 mmol/liter, the glucose excess is released into the urine, i.e. glucozuria sets in.
On the contrary, a decreased glucose percentage in the blood is known as hypoglycemia. Hypoglyce-
mia as low as about 1,5 mmol/liter leads to the syncopal state, while a still lower glucose concentra-
tion results in high excitability of the nervous system and ultimately leads to convulsing and coma.
Processes leading to hyperglycemia:
- absorption of glucose from the intestine (alimentary hyperglycemia);
- breakdown of glycogen to glucose (commonly, in liver)
- gluconeogenesis (in liver and kidney).
Processes leading to hypoglycemia:
- transport of glucose from the blood to tissue followed by glucose oxidation to end products;
- synthesis of glycogen from glucose in liver and skeletal muscles;
- production of triacylglycerol from glucose in fat tissue.
The dietary intake of carbohydrates leads to a short-term (within 1 or 2 hours) hyperglycemia
and, occasionally, glucosuria. The glucose level in the blood is monitored by neuron-hormonal nervo-
63
us system increases the glucose level in the blood, while excitation of the parasympathetic portion pro-
duces a reverse effect. The only hormone capable of reducing the glucose content is insulin. It stimula-
tes all of the three processes of glucose assimilation (intracellular transport and degradation of gluco-
se, synthesis of glycogen, and synthesis of triglyceride from glucose in fat tissue). All other hormones
make the glucose level increase; for this reason, they are occasionally referred to as contrainsular hor-
mones. These include adrenalin, glucagon, thyroxin and triidothyronin, somatotropin (which stimulate
glycogen degradation), and glucocorticoids (which stimulate gluconeogenesis).
Pathology of carbohydrate metabolism. Patients with diabetes mellitus type I have the deficiency
of active insulin, and patients with diabetes mellitus type II have the deficiency of number receptors
for insulin on the cell membranes. Patients with steroid diabetes mellitus have hyperproduction of
glucocorticoids (cortisol).
In diabetes mellitus take place the changes in carbohydrate, lipid, protein, and water-mineral me-
tabolisms. All patients with different types of these diseases have the character disturbances of meta-
bolism: hyperglycemia, glucosuria, ketonemia, ketonuria, hyperaminoacidemia and hyperaminoacidu-
ria, increased concentrations of fatty acids, glycerol, and cholesterol in blood. Insulin preparations are
used in the therapy of diabetes mellitus.
Enzymopathies of carbohydrate metabolism. Enzymopathies associated with an impaired glyco-
gen metabolism occur rather frequently. They manifest themselves either in glycogen accumulation in
the organs (accumulation diseases), or in lack of glycogen. The enzymopathies that lead to glycogen
deposition are called glycogenoses, and those, preventing glycogen deposition, aglycogenoses. Depen-
ding on the site of preferential glycogen deposition, three disease forms are distinguished: hepatic,
muscular, and generalized (Gierkes disease, Pompes disease, Hardys disease, etc.).
Galactosemia is a molecular disease evoked by a defective galactose-1-phosphate-uridyl transfe-
rase.This disease leads to the accumulation of galactose-1-phosphate, which, when in norm, rapidly
converts to uridine diphosphate galactose with subsequent involvement in the glucose conversion pat-
hway. Galactose-1-phosphate is toxic for the human organism. Since galactose (which makes part of
lactose) is consumed in large quantities by the infant from the breast milk, the organism of an infant
suffering from galactosemia rapidly accumulates this toxic galactose derivative. The infant loses in
weight, its mental and physical development becomes retarded, its liver increases in size, and the lenti-
cular opacity develops. If not withdrawn from the breast milk feeding, the infant usually dies from ali-
mentary toxicosis. In patients with galactosemia, galactose must be excluded from the diet.

LECTURE 14

Subject: LIPIDS: CLASSIFICATION, CHEMICAL STRUCTURE AND
DIGESTION OF LIPIDS

Lipids are organic compounds of biological nature, insoluble in water but soluble in nonpolar sol-
vents such as chloroform, ether, or benzene. The main features enabling assignment of a compound to
the lipid class are: biological origin, hydrophobicity, solubility in nonpolar liquids and insolubility in
water, occurrence of higher alkyl radicals or carbohydrates.
Classification of lipids. A number of classifications have been proposed for lipids on the basis of
their structural, physico-chemical, biological, or physiological properties. The most sophisticated clas-
sification appears to be the structural one, which takes into account structural features of lipid com-
pounds.
Simple molecules:
- glycerides (acylglycerides),
- waxes,
- steroids.
Complex lipids (heterolipids):
- phospholipids,
- glycolipids,
- sphingolipids.
The differentiation of lipids according to their physico-chemical properties takes into account the
extent of polarity exhibited by the lipid molecules. By this property, the lipids are classified into neu-
tral (or nonpolar) and polar species. Lipids possessing no charge belong to the former type. The latter
64
type includes charged lipids exhibiting distinct polar properties (for example, phospholipids and fatty
acids).
According to their physiological importance, the lipids are divided into reserve lipids and structu-
ral lipids. The reserve lipids are stored in large amounts and then consumed to supply the energetic re-
quirements of the organism. Acylglycerides rank among reserve lipids. The former are primarily used
in the buildup of biological membranes, covering (protective) layers in plants, insects, and skin in ver-
tebrates.
Lipids account for about 10-20% of the total mass of the human organism. The body of an adult
human contains on the average 10 to 12 kg of lipids, of which the structural lipids (major constituents
of biomembranes) account for 2 or 3 kg; the rest are reserve lipids. About 98% of the latter are deposi-
ted in adipose tissues. Among the tissues of the organism, the structural lipids are distributed nonuni-
formly. The nerve tissue is the most rich one is structural lipids (to 20-25%). In biomembranes of the
cell, the lipids make up about 40% (by dry weight).
Simple lipids
Glycerides, or acylglycerides, constitute the most widespread group of simple lipids. By their
chemical structure, they are esters of fatty acids with a tribasic alcohol, glycerol. Glycerides (acylgly-
cerides) are also called neutral lipids because of their rather inert character. Glycerides are divided into
mono-, di-, and triacylglycerides containing one, two, and three acyl groups (R-CO-), respectively:


Figure 39. Chemical structures of glycerol and triglycerides

The name for a neutral lipid is derived from the name of the constituent fatty acid and the ending
glyceride. For example, palmitoylglyceride is a monoglyceride containing a palmitic acid moiety;
tripalmitoylglyceride is a triglyceride containing three identical acyl residues.
Differentiated are simple acylglycerides containing residues of the same fatty acid (as typified by
the above case), and complex acylglycerides composed of residues of two or three different fatty
acids. Natural neutral lipids are mixtures of simple and complex glycerides with a prevalence of unsa-
turated triglycerides.
CH
3
(CH
2
)
4
CH=CHCH
2
CH=CH(CH
2
)
7
COOH
Linoleic acid
CH
3
(CH
2
)
4
CH=CHCH
2
CH=CHCH
2
CH=CHCH
2
CH=CH(CH
2
)
3
COOH
Arachidonic acid
CH
3
(CH
2
)
7
CH=CH(CH
2
)
7
COOH
Oleic acid
Physico-chemical properties of neutral lipids are largely determined by their composition. The
melting points are higher in acylglycerides containing higher saturated acyl groups. Saturated fatty
acids (for example, tristearin) are found, as a rule, in solid fats, and in liquid fats, unsaturated acids
(for example, triolein). All acylglycerides are less dense than water. They are soluble in nonpolar sol-
vents (chloroform, benzene, ether, and hot ethanol). Only mono- and diacylglycerides containing free
polar hydroxyl groups are water-soluble. They form micelles in water. Triacylglycerides are insoluble
in water and are incapable of micellation. When subjected to basic hydrolysis or saponification, acyl-
glycerides regenerate glycerol and free fatty acids. In the organism, acylglycerides are hydrolyzed by
special enzymes, lipases.
Waxes are mixtures of ethers and esters derived from higher monobasic alcohols and higher fatty
acids. High hydrophobicity of waxes form a waterrepellent protective layer (grease) on the leaves and
fruit of plants, skin and hair of animals, feathers of birds, and the external skeleton of insects. They
constitute part of honey wax.
Sterids are also known as of frequent occurrence. They are contained in products of animal origin
(butter and egg yolk). In human and animal organisms, most of the tissue cholesterol (about 60-70%)
occurs as cholesterol esters. In blood, cholesterol esters as constituents of transport lipoproteins make
up the major content of the overall cholesterol. It is quite probable that cholesterol esters provide for a
specific form of storing cholesterol in tissues.
65
Lanolin (wool fat) is also of sterid origin (a mixture of fatty acid esters of lanosterol and agnoste-
rol) and is widely used in the pharmaceutical industry as an ointment base. Sterids such as fatty acid
esters of stigmasterol, ergosterol, and |-sitosterol constitute a significant part of overall sterols inpla-
nts.

Figure 40. Chemical structure of cholesterol

Cholesterol has been classified as a structural lipid. It forms part of biological membranes of the
cells with a predominant content in the plasmatic (cellular) membrane rather than in other membranes
(those of mitochondria, microsomes, nuclei, etc.).
Among the steroid compounds of animal and plant origin, the following biologically active cho-
lesterol derivatives should be mentioned bile alcohols, bile acids, steroid hormones, D vitamins, ste-
roid glycosides, steroid alkaloids. The most widespread bile acids in the human bile are cholic and
chenodeoxycholic acids.











Figure 41. Chemical structure of bile acid: glycocholic acid

In liver these acids react easily with glycine and taurine to form coupled compounds, or conjuga-
tes, for example, glycocholic and taurocholic acids.
In the intestine and bile, the bile acids occur as anions (bile salts). Viewed structurally, the bile
acids are amphipathic molecules, since they possess a hydrophobic ring and hydrophilic groups, viz.
hydroxyl properties and favour thereby the emulgation of lipids in the intestine. The bile acids are
essential for normal digestion and absorption of lipids in the intestine. They also assist in the absorp-
tion of fat-soluble vitamins in the digestive tract. Sparing solubility of bile acids is the reason for the
formation of gallstones (in humans, the bile acids account for about 1% of the gallstone composition).
Complex lipids or heterolipids
Complex lipids (heterolipids), as distinct from simple lipids, contain a nonlipid component, which
may be a phosphate, carbohydrate, etc. According to their chemical structure, the representatives of
complex lipids, i.e. phospholipids, glycolipids and sphingolipids, belong to molecular species with a
higher level of structural organization.









66

















Figure 42. Phospholipid structure

Phospholipids are complex representative of phosphate-substituted esters of diverse organic alco-
hols (glycerol, sphingosines, and diols). All of the phospholipids are polar lipids and are predominan-
tly contained in the cell membranes. In the fat depots, their occurrence is insignificant.
Phosphoglycerides. In phosphoglycerides, one of the hydroxyl groups forms an ester bond with a
phosphate, rather than with a fatty acid.
Phosphatidic acid is the simplest representative of naturally occurring phosphoglycerides. Phos-
phatidylethanolamines and phosphatidylcholines are very widespread phosphoglycerides present in the
membranes of all types. They account for 20% and 50% of the cell membrane lipids. Plasmalogens are
especially abundant in the brain and spinal marrow, constituting 50-90% of the whole of lipids. A spe-
cific feature of plasmalogen structure is the formation of an ether bond with a higher aliphatic aldehy-
de.
Glycolipids are complex lipids containing a carbohydrate component. They include the simplest
glycolipids, glycosyldiacylglycerols, in which one hydroxyl group of the glycerol moiety is replaced
by a monosaccharide. Glycoshpingolipids occur in the nerve cells where they appear to be essential for
the normal electric activity and transmittance of nervous impulses. Cerebrosides, gangliosides, and
sulpholipids belong to these lipids.
Cerebrosides were first isolated from the brain (cerebrum), to which they owe their name. Their
carbohydrate moiety is most commonly represented by galactose or, very rarely, by glucose. Fatty
acids forming part of cerebrosides are quite numerous and include lignoceric (n-tetracosanoic), cere-
bronic (2-hydroxytetracosanoic), nervonic, and hydroxynervonic fatty acids.
Sulpholipids are sulphated derivatives of cerebrosides. The sulphate moiety is added to the third
hydroxyl group of galactose, sulpholipids exhibit pronounced acidic properties and are capable of
easily binding cations. Presumably, they participate in the cation transport across the membranes of
nervous cells and fibers. Therefore, sulpholipids are necessary for the normal electrical activity of the
nervous system.
Gangliosides, in contrast to other glycosphingolipids, contain an olygosaccharide composed of
different monosaccharides. Ganglioside components are diversified and their masses vary over a wide
range. The cerebral cortex cells are rich in gangliosides.
Sphingolipids (sphingophosphatides). In the group of sphingolipids, sphingosine is the alcohol
entity. Its N-acylated derivative, ceramide, is the ancestor for all of the sphingolipids. Sphingolipids
are divided into sphingomyelins, or sphingophosphatides, and glycosphingolipids.They differ in that
the sphingomielins, like all phospholipids, have the hydroxyl group substituted by a phosphate or a
residual alcohol (mostly choline), while glycolipids bear, in place of the phosphate group, a monosac-
charide (in cerebrosides) or an oligosaccharide (in gangliosides) containing galactose and a number of
aminosaccharides.
Sphingomyelins are contained in the nerve tissue in large amounts and form part of myelin which
serves as a material for the nerve fibre sheath. They have been found in the lung, liver, kidney, spleen,
blood, and other organs. A special period, the so-called myelinization period, is singled out during the
67
human and animal brain development. In man, the myelinization period, i.e. the formation of sphingo-
myelin in the nerve sheath and nervous system, starts immediately after the birth and continues for
about four months.
In large amounts sphingomyelins are found in the blood plasma and the erythrocyte envelope: 8-
15% and 30-40% of the total lipid mass, respectively. This appears to be associated with the function
of erythrocytic membranes.
Biological functions of lipids are determined by their structural and physicochemical properties.
Digestion mechanism for lipids
Digestion of lipids takes place in the sections of digestive tract where the following necessary
conditions have been provided for:
1. Availability of lipolytic enzymes capable of hydrolyzing lipids.
2. Conditions providing for emulsifying lipids.
3. Optimal pH of medium for the action of lipolytic enzymes (the medium should be neutral or
weakly alkaline).
All of these conditions are provided for in the intestine of adult human. In infants, especially new-
borns, similar conditions are created for digesting milk triacylglycerides by gastric lipase. The medium
pH for the infants stomach contents is about 5,0 (weakly acidic medium), and milk fat occurs in an
emulsified state; therefore fat is amenable to cleavage by lipase to a certain extent. In the adult man, a
strongly acidic medium inactivates the gastric lipase.
In the intestine, the food delivered from the stomach becomes neutralized, and the fat undergoes
emulsification. Emulsification of lipids is effected by bile acids supplied to the intestine as bile com-
ponents. The major bile acids contained in the bile are: cholic and chenodeoxycholic acids and their
glycine and taurine derivatives: glycocholic and taurochenodeoxycholic acids.
Bile acids perform biological functions:
- as emulsifying agents;
- as activators of lipolytic enzymes;
- as transport means, since, by forming a transport complex with fatty acids, bile acids facilitate
the intestinal absorption of the former. All bile acids are amphiphilic compounds and therefore exhibit
emulsifying properties.
Bile acid molecules adsorbed at the fat/water interface prevent the phase separation. The intesti-
nal peristalsis facilitates dispersion of large fat droplets, while the bile acid molecules retain finely dis-
persed fat particles on a suspended state and prevent their coalescence. Free fatty acids and monoacyl-
glycerides produced by digestion of lipids, dietary phospholipids, and their partly digested products
(phosphatidylcholine) are auxiliary emulsifying agents.
Hydrolysis of triacylglycerides which constitute the major mass of dietary lipids is effected by the
action of pancreatic lipase. The lipase is supplied in an inactive form. In the intestine, it is activated
by a special cofactor, colipase, and by bile acids. The activated lipase acts on the triacylglycerides of a




Action of bile salts in digestion
suspended fat droplet. The enzyme itself is dissolved in the aqueous phase; nonetheless, it splits a sub-
strate found in the lipid phase. Lipase possesses a special hydrophobic site (head) which comes into
contact with triacylglyceride. The hydrolyzates are mostly 2-monoacylglyceride and free fatty acids.

68


Figure 43. Reaction of TG hydrolysis by lipase

The intestinal and pancreatic juice carboxyesterases split 2-monoacylglyceride into a free acid
and glycerol. The hydrolysis of triacylglycerides is assisted by calcium ions that form complexes with
free fatty acids.
Hydrolysis of phospholipids is carried out by a group of lipolytic enzymes called phospholipases.
A variety of phospholipases are known, denoted A
1
, A
2
, C and D. They hydrolyze various bonds in the
phospholipid molecule, as exemplified by phosphatidylcholine.
Activation of phospholipase A
2
occurs in the intestinal juice, via splitting a hexapeptide from the
proenzyme by trypsin. Besides, the intervention of bile acids and calcium ions is needed for the activi-
ty of phospholipase A
2
and other phospholipases. Bile acids assist in the approach of a substrate to the
active centre of enzyme, while calcium ions remove free fatty acids from the site of enzymic activity
(as in the case of lipase) and prevent inactivation of phospholipase.
The action of phospholipase A
2
, which is the chief digestive phospholipase, results in the formati-
on of extremely toxic lysophosphatides that are immediately hydrolyzed by lysophospholipase.
The end products of hydrolysis are glycerol, fatty acids, an inorganic phosphate and a residual al-
cohol (choline, ethanolamine, inositol, or serine).
Hydrolysis of steroid esters. Dietary cholesterol esters which are abundant in certain food pro-
ducts (egg yolk, butterfat, caviar, etc.) are hydrolyzed in the emulsion droplets of intestinal contents by
pancreatic cholesterolesterase. This enzyme is also activated by bile acids. The enzymic hydrolysis
leads to free cholesterol and fatty acids. The hydrolyzates of all dietary lipids are absorbed in the intes-
tine.
The uptake of lipid hydrolyzates and their transport. The absorption of digested lipid products
exhibits a number of specific features. For example, the uptake of fatty acids depends on the carbon
chain length. Short-chain fatty acids (to 10-12 carbon atoms) are transported into the intestinal epithe-
lium by simple diffusion. Long-chain fatty acids (over 14 carbon atoms) form transport complexes
with bile acids. The complexes thus formed are called choleinic acids. Therefore, the fatty acids pass
across the intestinal epithelium membrane in a complexed state. This kind of transport may be regar-
ded as a facilitated transport in which bile acids act as carriers. Within the intestinal wall, the choleinic
complexes dissociate, and the bile acids are excreted into the blood of portal vein and transferred to
the liver. From the liver, the bile acids return with bile to the intestine. This turnover is referred to as
the hepatoenteric circulation of bile acids.
Besides, easily absorbed are glycerol, phosphates (as sodium and potassium salts), choline and
other alcohols, sphingosine and cholesterol.
Glycerol and fatty acids are the starting materials for the resynthesis of triacylglycerides. The rest
of phosholipids, all the triacylglycerides, free and esterified cholesterol are transported in the lymph.
The intestinally resynthetized lipids are transported as constituents of chylomicrons. From the thoracic
69
lymphatic duct, chylomicrons are supplied into the blood which becomes milky turbid and distinctly
opalescent (such a blood plasma is called lipemic). In blood, chylomicrons or more exactly, their cons-
tituent triacylglycerides are split by lipoprotein lipase. This enzyme hydrolyzes the chylomicron tria-
cylglycerides into glycerol and fatty acids.

LECTURE 15

Subject: CATABOLISM OF LIPIDS IN TISSUES

Hydrolysis of triacylglycerides in tissues is effected by a tissue enzyme, triacylglyceride lipase,
which hydrolyzes triacylglycerides to glycerol and free fatty acids. There are a variety of tissue lipases
that differ primarily in their optimum pH and their location in the cell. The acidic lipase is contained in
lysosomes; the basic lipase, in microsomes; and the neutral lipase, in cytoplasm. A specific feature of
the tissue lipase is its sensitivity to hormones which, by activating adenylate cyclase, elicit the transiti-
on of the inactive tissue lipase to its active form via phosphorylation with protein kinase. Lipases mo-
bilize triacylglycerides. This process is also known as the tissue lipolysis.
The cell membrane phosphoglycerides are hydrolyzed with phospholipases A
1
, A
2
, C, and D
which are located chiefly in lysosomes. In the adipose tissue, glycerol and fatty acids as produced by
triglyceride hydrolysis are not subject to oxidation and are released into the blood to be supplied to
other organs.

Oxidation of glycerol
The glycerol metabolism is closely related to the glycolysis involving glycerol metabolites accor-
ding to the following scheme:

H
2
C-OH H
2
C-OH H
2
C-OH
| Glycerol | o-Glycerol phosphate |
HC-OH phosphokinase HC-OH dehydrogenase C=O
| | |
H
2
C-OH ATPADP H
2
C-OPO
3
H
2
NADNADH
+
+H
+
H
2
C-OPO
3
H
2

glycerol glycerol phosphate dihydroxyacetone
phosphate

At first, glycerol converted to glycerol phosphate through the agency of glycerol phosphokinase.
Glycerol phosphate, by action of NAD-dependent glycerol phosphate dehydrogenase, is converted to
dihydroxyacetone phosphate, which, as a common glycolysis metabolite, enters glycolysis to be redu-
ced by enzymes to lactate under anaerobic conditions, or to CO
2
and H
2
O under aerobic conditions.
Conversion of one glycerol molecule yields one ATP molecule under anaerobic and 22 ATP molecu-
les, under aerobic conditions. Glycerol is a profitable energy substrate and it used as an energy source
practically by all organs and tissues.

Oxidation of fatty acids
Oxidation of higher fatty acids was first studied in 1904 by Knoop. Knoop coined the fatty acid
oxidation mechanism as |-oxidation.
The fatty acids, as produced by intracellular hydrolysis of triacylglycerides or supplied to the cell
from the blood, must be brought into a state of activation. Their activation is effected in the cytoplasm
with the participation of acyl-coA synthetase:

acyl-coA synthetase
R-CH
2
-CH
2
-COOH+ATP + coA-SH R-CH
2
-CH
2
-CO~ScoA+ AMP +P

Since the activation process is effected extramitochondrially, transport of acyls across the mem-
brane into the mitochondria is necessary. The transport is accomplished with the participation of carn-
itine, which takes up the acyl from acyl-coA on the outer membrane side. Acylcarnitine assisted by
carnitine translocase diffuses to the inner side of the membrane to give its acyl to the HScoA located
in the matrix. The process of reversible acyl transfer between HScoA and carnitine on the outer and in-
ner sides of the membrane is effected by the enzyme acyl-coA carnitine transferase.
70

















Figure 44. Fatty acid transport into mitochondrium

The |-oxidation of fatty acids occurs in the matrix of mitochondrium.







71

Figure 45. Scheme of fatty acid oxidation

Each turn, or revolution, of the fatty acid spiral produces an acetic acid residue split from the
fatty acid as acetyl-coA to yield one FADH
2
molecule and one NADH
2
molecule. The cycle turns are
then repeated until the fatty acid chain becomes shortened to a four-carbon fragment, i.e. butyryl-coA.
In the last turn, butyryl-coA splits apart, and two, rather than one, acetyl-ScoA molecules are formed:

CH
3
-CH
2
-CH
2
-CO~ScoA
. \
CH
3
-CO~ScoA + CH
3
-CO~ScoA

The oxidation products of an even-numbered fatty acid are acetyl-coA, FADH
2
and NADH
2
. Sub-
sequently, acetyl-coA enters the Krebs cycle, and FADH
2
and NADH
2
are directly supplied to the res-
piratory chain.
Energy balance of fatty acid oxidation
The energetic value of an even-numbered fatty acid is estimated in the following manner. Com-
plete oxidation of a fatty acid composed of n carbon atoms yields (n2) acetyl-ScoA molecules (each
acetyl containing two carbon atoms) and [(n2) 1] FADH
2
and NADH
2
molecules (since the last
turn of the fatty acid spiral yields two acetyl-coSA molecules, one FADH
2
molecule and one
NADH
2
molecule). Oxidation of FADH
2
gives two ATP molecules, and oxidation of NADH
2
, three
ATP molecules, i.e. a total of 5 ATP molecules, or, in the general case, 5[(n2 1] ATP molecules. As
has been noted above, the complete oxidation of one acetyl-ScoA molecule results in the formation of
(12 n2) ATP molecules. One ATP molecule being used for the fatty acid activation, [(12n2) 1]
ATP molecules remain. For example, the palmitic acid molecule, which contains 16 carbon atoms,
produces 130 ATP molecules.
The energetic value of fatty acids is superior, for example, to that of glucose. For example, the
complete oxidation of capronic acid (whose molecule contains the same number of carbon atoms as
glucose) yields 45 ATP molecules as compared with 38 molecules which can be derived from glucose.
However, the acetyl-ScoA molecules as produced by |-oxidation require a sufficient amount of oxalo-
acetate to be degraded by the Krebs cycle. In this respect, carbohydrates have an advantage over fatty
acids, since the breakdown of the former species leads to pyruvate serving as a source for both acetyl-
coA and oxaloacetate (pyruvate-carboxylase reaction), i.e. the acetyl-coA conversion within the Krebs
cycle is thus facilitated. It is not without reason that in the older biochemical literature the notion that
fats burn down in the carbohydrate flame was popular, since the ATP from glycolysis can be used
for the cytoplasmic activation of fatty acids, while the pyruvate-derived oxaloacetate facilitates the in-
sertion of fatty acid acetyl residues into the Krebs cycle.
The specific behaviour of odd-numbered fatty acids under oxidation is that one propionyl-ScoA
molecule (CH
3
-CH
2
-CO~ScoA) per oxidized fatty acid molecule, alongside the products acetyl-coA,
FADH
2
, and NADH
2
(common to even-numbered fatty acids), is formed. Propionyl-ScoA converts to
succinyl-ScoA:
CO
2

+
CH
3
-CH
2
-CO~ScoA CH
3
-CH-CO~ScoAHOOC-CH
2
-CH
2
CO~ScoA
ATPADP |
COOH

propionyl-ScoA methylmalonyl-ScoA succinyl-ScoA

Succinyl-ScoA enters the Krebs cycle.
The specific behaviour of unsaturated fatty acids under oxidation is determined by the position
and the number of double bonds in the fatty acid molecule. The stepwise oxidation of an unsaturated
acid to the position of a double bond in it proceeds in a manner similar to that of saturated acid oxida-
tion. The oxidation reaction proceeds with the involvement of an accessory enzyme, 3,4-cis-2,3-trans-
72
enoyl-ScoA isomerase; this facilitates the translocation of the double bond to an appropriate position
and alters the double-bond configuration from cis to trans.

LECTURE 16

Subject: BIOSYNTHESIS OF LIPIDS IN TISSUES. REGULATION AND
PATHOLOGY OF LIPID METABOLISM

Biosynthesis of fatty acids. In the organism tissues, fatty acids are continually renewed in order
to provide not only for the energy requirements, but also for the synthesis of multicomponent lipids
(triacylglycerides, phospholipids, etc.). In the organism cells, fatty acids are resynthetized from sim-
pler compounds through the aid of a supramolecular multienzyme complex referred to as fatty acid
synthetase. Since in mammals palmitic acid in this process is a major product, this multienzyme com-
plex is also called palmitate synthetase. Biosynthesis of fatty acids exhibits a number of specific featu-
res:
1. Fatty acid biosynthesis, being distinct from oxidation, is localized in the endoplasmic reticu-
lum.
2. The source for the synthesis is malonyl-ScoA, which is produced from acetyl-ScoA.
3. Acetyl-ScoA is involved in the synthetic reaction as a primer only.
4. NADPH
2
is used to reduce fatty acid biosynthesis intermediates.
5. All the steps of malonyl-ScoA fatty acid biosynthesis are cyclic processes that occur on the
surface of palmitate synthetase.
Production of malonyl-ScoA for fatty acid biosynthesis. Acetyl-ScoA serves as a substrate in the
production of malonyl-ScoA. There are several routes by which acetyl-coA is supplied to the cyto-
plasm. One route is the transfer of acetyl residues from the mitochondrial matrix across the mitochon-
drial membrane into the cytoplasm. This process resembles a fatty acid transport and is likewise effec-
ted with the participation of carnitine and the enzyme acetyl-ScoA-carnitine transferase. Another route
is the production of acetyl-ScoA from citrate. Citrate is delivered from the mitochondria and undergo-
es cleavage in the cytoplasm by the action of the enzyme ATP-citrate lyase:

Citrate + ATP + HScoA acetyl-ScoA + oxaloacetate + ADP + H
3
PO
4


The acetyl-ScoA supplied to the cytoplasm via the above routes is used for the synthesis of malo-
nyl-ScoA:
CH
3
COOH
|

|

C=O + HCO
3
+ ATP CH
2
+ ATP + H
3
PO
4

\ |
ScoA C=O
\ ScoA
Steps of fatty acid biosynthesis assisted by palmitate synthetase. The acyl carrier protein (ACP)
is located at the centre of the multienzyme complex. ACP acts both as an acceptor and a distributor of
acyl groups. The cyclic process of fatty acid synthesis may be represented by a series of consecutive
reactions:




73


Figure 46. Scheme of fatty acid biosynthesis

The synthetic cycle is thus repeated. For example, seven cycles are implicated in palmitc acid bio-
synthesis and, accordingly, seven malonyl residues and one acetyl are required.
Fatty acid chain elongation. In the mitochondria, the chain elongation is achieved through the
aid of an enzyme complex by adding acetyl residues from acetyl-ScoA. In the endoplasmic reticulum,
the chain elongation is accomplished by an enzyme complex through making use of malonyl-ScoA.

Biosynthesis of unsaturated fatty acids
In the mammalian tissues, the formation of monoene fatty acids is only possible. Oleic acid is
derived from stearic acid, and palmitooleic acid, from palmitic acid. This synthesis is carried out in the
endoplasmic reticulum of the liver cells via the monooxigenase oxidation chain. Any other unsaturated
fatty acids are not produced in the human organism and must be supplied in vegetable food. Polyene
fatty acids are essential food factors for mammals (vitamin like substances).

Biosynthesis of triglycerides
Triglyceride biosynthesis proceeds with the involvement of the lipids deposited in fat tissue or in
other tissues of the organism. This process is localized in the hyaloplasm of cells.
o-Glycerol phosphate and acyl-ScoA, rather than corresponding free glycerol and free fatty acid,
are utilized in the direct synthesis of triglycerides. o-Glycerol phosphate is produced either by phos-
phorylating the glycerol supplied to the tissue, or by reducing dihydroxyacetone phosphate as an inter-
mediary product of glycolysis.
CH
2
-OH CH
2
-OH
| glycerol kinase | R
1
-CO~ScoA glycerophosphate
HC-OH HC-OH + acyltransferase
| ATPADP | R
2
-CO~ScoA
CH
2
-OH CH
2
-OPO
3
H
2
- 2coASH

o-glycerol o-glycerol phosphate

H
2
C-O-CO-R
1
H
2
C-O-CO-R
1
H
2
C-O-CO-R
1
| phosphotidate | diglyceride |
HC-O-CO-R
2
phosphotase HC-O-CO-R
2
acyltransferase HC-O-CO-R
2

| | |
H
2
C-OPO
3
H
2
-H
3
PO
4
H
2
C-OH + R
3
-CO~ScoA H
2
C-O-CO-R
3

phosphatidic acid diglyceride triglyceride
Phospholipid biosynthesis
Biosynthesis of phospholipids is associated with the renewal of membranes. This process is
accomplished in the tissue hyaloplasm. The first steps of phospholipid and triglyceride biosyntheses
coincide; subsequently, these routes diverge at the level of phosphatidic acid and diglyceride. Two
routes of phospholipid biosynthesis are known; in either, the participation of CTP is necessary.
74

Figure 47. Scheme of phospholipid synthesis

The first route involves phosphatidic acid in phosphoglyceride biosynthesis. Phosphatidic acid
reacts with CTP to yield CDP-diglyceride which, as a coenzyme, can participate in the transfer of dig-
lyceride onto serine (or inositol) to produce phosphatidylserine (or phosphatidylinositol). Serine phos-
phatides are liable to decarboxylation (pyridoxal phosphate acting as a coenzyme) to yield ethanolami-
ne phosphatides. The latter species are subject to methylation by S-adenosylmethionine (which dona-
tes three methyl groups), tetrahydrofolic acid and methylcobalamine acting as methyl group carriers.
The second synthetic route involves activation of an alcohol (for example, choline) to produce
CDP-choline. The latter participates in the transfer of choline onto diglyceride to form phosphatidyl-
choline.
The phospholipids thus obtained are transported by lipid-carrier cytoplasmic proteins to the mem-
branes (cellular or intracellular) to replace the used or impaired phospholipid molecules.
Because of the competition between the phospholipid and triglyceride synthetic routes for com-
mon substrates, all substances that favour the former route impede the tissue deposition of triglyceri-
des. Such substances are referred to as lipotropic factors. They include: choline, inositol, and serine, as
structural components of phospholipids; pyridoxal phosphate, as an agent facilitating the decarboxyla-
tion of serine phosphatides; methionine, as a donor of methyl group transfer coenzymes (tetrahydrofo-
lic acid and methylcobalamine). They may be used as drugs preventing excessive deposition of trigly-
cerides in tissues (the so-called fatty infiltration).
Biosynthesis of ketone bodies
Three compounds: acetoacetate, |-hydroxybutirate, and acetone, are known as ketone bodies.
They are suboxidized metabolic intermediates, chiefly those of fatty acids and of the carbon skeletons
of the so-called ketogenic amino acids (leucine, isoleucine, lysine, phenylalanine, tyrosine, and trypto-
phan). The ketone body production, or ketogenesis, is effected in the hepatic mitochondria (in other
tissues, ketogenesis is inoperative). Acetyl-ScoA is the starting compound for the biosynthesis of keto-
ne bodies.
CH
3
-C~ScoA + CH
3
-C~ScoA CH
3
-C-CH
2
-C~ScoA + coASH +
|| || || ||
O O O O
acetyl-ScoA acetyl-ScoA acetoacetyl-ScoA

CH
3

75
|
CH
3
-C~ScoA HOOC-CH
2
-C-CH
2
-C~ScoA
|| - coASH | ||
O OH O
acetyl-ScoA |-hydroxy-|-methylglutaryl-ScoA

CH
3
-C~ScoA + HOOC-CH
2
-C-CH
3

|| ||
O O
acetyl-ScoA acetoacetate
O OH
|| |
HOOC-CH
2
-C-CH
3
HOOC-CH
2
-CH-CH
3
+ -CO
2
NADH
+
+ H
+
NAD |-hydroxybutyrate
H
3
C-C-CH
3
||
O
acetone

Biosynthesis of cholesterol
Biosynthesis of cholesterol from acetyl-ScoA proceeds as assisted by the enzymes of endoplas-
mic reticulum and hyaloplasm, in many tissues and organs. This process is especially active in the li-
ver of adult humans. Cholesterol biosynthesis is a multistage process; in general, it may be divided in-
to three steps;
1. Production of mevalonic acid from acetyl-ScoA.
2. Synthesis of an active isoprene from mevalonic acid followed by the condensation of the
former to squalene.
3. Conversion of squalene to cholesterol.
The initial reactions in the first step, prior to the formation of |-hydroxy-|-methylglutaryl-ScoA,
resemble those involved in ketogenesis with the only distinction that ketogenesis occurs in the mi-
tochondria, while cholesterol biosynthesis is carried out extramitochondrially:

2 Acetyl-ScoA acetoacetyl-ScoA + acetyl-ScoA
|-hydroxy-|-methylglutaryl-ScoA

Further, |-hydroxy-|-methylglutaryl-ScoA is converted with hydroxymethylglutaryl-ScoA into
mevalonic acid:
CH
3
CH
3

| |
HOOC-CH
2
-C-CH
2
-C~ScoAHOOC-CH
2
-C-CH
2
-CH
2
OH+coASH
| || +2NADPH
2
|
OH O 2NADP OH

This reaction is irreversible and is a rate-limiting stage of the overall cholesterol biosynthesis.


Cholesterol

During the second step, the two compounds constitute the active isoprene, which is consumed
in the production of squalene: squalene lanosterin cholesterol
Cholesterol, mostly esterified, is utilized in the buildup of cell biomembranes. Besides, choleste-
rol is a precursor of biologically important steroid compounds: bile acids (in liver), steroid hormones
76
(in adrenal cortex, male and female sexual glands, and placenta), and vitamin D
3
, or cholecalciferol (in
skin).


Cholecalciferol

Prostaglandins are hormone-like compounds (hormonoids) derived from C
20
-polyene fatty acids
containing a cyclopentane ring. They are short-lived species that are synthetized at need in small
amounts to exert a local biological effect at the site of their formation.


Prostaglandin E
1
(left) and thromboxane A
2


Figure 48. Prostaglandines: pathways in biosynthesis of eicosanoids from arachidonic acid

Regulation of lipid metabolism in the organism
The rate of lipid metabolism in the organism tissues is dependent on the dietary supply of lipids
and on the neurohormonal regulation. An excessive intake of high-calory food (carbohydrates and tri-
glycerides) impedes the consumption of endogenic triglyceride reserves stored in the fat tissues. More-
over, carbohydrates provide a very favourable basis for the neogenesis of various lipids; for this rea-
77
son, a large dietary intake of carbohydrate-rich food exerts a significant influence on the production of
triglycerides and cholesterol in the organism.
Synthesis of endogenic cholesterol is also controlled by exogenous cholesterol supplied in food:
the more dietary cholesterol is digested, the less endogenic cholesterol is produced in the liver. Exoge-
nous cholesterol inhibits the activity of hydroxymethylglutaryl-ScoA reductase and the cyclization of
squalene to lanosterol.
The dietary ratio of various lipids plays an important role in the lipid metabolism in the organism.
The available amounts of polyene fatty acids and phospholipids acting as solvents for fat-soluble vita-
mins affect not only the absorption of the latter species, but also the solubility and stability of choleste-
rol in the organism fluids (blood plasma and lymph) and biliary ducts. Vegetable oils with a high per-
centage of phospholipids and polyene fatty acids impede an excessive accumulation of cholesterol and
its deposition in blood vessels and other tissues, and facilitate the removal of cholesterol excess from
the organism. Polyene fatty acids are needed in the production of prostaglandins.
The lipotropic factors exercise a marked effect on the biosynthesis of phospholipids and triglyce-
rides. The dietary deficiency of lipotropic factors favours the triglyceride production in the organism.
The neuro-hormonal control of lipid metabolism chiefly affects the mobolization and synthesis of
triglycerides in the fat tissue. The lypolysis in tissues is dependent upon the activity of triglyceride
lipase. All the regulators that favour the conversion of the inactive (nonphosphorylated) lipase to the
active (phosphorylated) one stimulate the lipolysis and the release of fatty acids into the blood. Adre-
nalin and noradrenalin (secreted in the sympathetic nerve endings) hormones, (glucagon, adrenalin,
thyroxin, triiodothyronine, somatotropin, |-lipotropin, corticotropin, etc.), tissue hormones, including
biogenic amines (histamine, serotonin, etc.), act as stimulators for this process. Insulin, on the contra-
ry, inhibits the adenylate cyclase activity; preventing thereby the formation of active lipase in the fat
tissue, i.e. retards the lipolysis. In addition, insulin favours the neogenesis of triglycerides from carbo-
hydrates, which, on the whole, provides for lipid deposition in the fat tissues as well as for the choles-
terol production in other tissues. The thyroid hormones thyroxin and triiodthyronine assist in the oxi-
dation of the cholesterol side chain and in the biliary excretion of cholesterol in the intestine.

Pathology of lipid metabolism
Most commonly, the lipid metabolism pathology is manifest as hyperlipemia (elevated concentra-
tion of lipids in blood) and tissue lipidoses (excessive lipid deposition in tissues). Normally, the lipid
contents in the blood plasma are:
- total lipids: 4-8 g/L
- triglycerides: 0,5-2,1 mmol/L
- total phospholipids: 2,0-3,5 mmol/L
- total cholesterol: 4,0-9,8 mmol/L (esterified cholesterol accounts for 2/3 of total cholesterol).
Hyperlipemia may manifest itself by an increased concentration of lipids, or certain groups the-
reof. For example, hypercholesterolemia and hypertriglyceridemia may be mentioned in this connec-
tion. Since practically all the blood plasma lipids make part of lipoproteins, hyperlipemias may be re-
duced to one of the hyperlipoproteinemia forms which differ in the varied rations of plasma lipopro-
teins of different groups.
Hyperlipoproteinemia is characterized by the enhanced content of chylomicrons in the blood
plasma; simultaneously, the percentage of o- and |-lipoproteins may be lowered. The triglyceride
content is 8-10 times above the norm, while cholesterol does not exceed the normal level.
Secondary hyperlipoproteinemias, which arise from a disordered lipid tissue metabolism or its
impaired control, are observed in diabetes mellitus, thyroid gland hypofunction, alcoholism, etc.
Tissue lipidoses. Hyperlipoproteinemias may lead to tissue lipidoses and also arise from heredita-
ry defects of the enzymes involved in the synthesis and breakdown of lipids in the tissues.
Atherosclerosis is a wide-spread pathology, manifested chiefly by the deposition of cholesterol in
arterial walls, which results in the formation of lipids plaques (atheromas). Lipid plaques are specific
foreign bodies around which the connective tissue develops abnormally (this process is called sclero-
sis). |-Lipoproteins and, partly, pre-|-lipoproteins containing much cholesterol exhibit atherogenic
properties.
Fatty infiltration of the liver. In this pathology, the triglyceride concentration in the liver is 10-
fold superior to the norm. The accumulation of fat in the cytoplasm is numerous; one of these may be
a deficiency in lipotropic factors and the associated therewith synthesis of excess of triglycerides.
78
Ketosis is a pathologic state produced by an excess of ketone bodies in the organism. Ketosis may
be regarded as lipid metabolism pathology with a certain reserve, since excessive biosynthesis of keto-
ne bodies in the liver is sequent upon an intensive hepatic oxidation not only of fatty acids, but also of
ketogenic amino acids. The breakdown of carbon frameworks of these amino acids leads to the forma-
tion of acetyl-ScoA and acetoacetyl-ScoA, which are used in ketogenesis. The ketosis is accompanied
by ketonemia and ketonuria, which is manifested by the increased concentration of ketone bodies in
blood and their excretion in the urine. In an aggravated form of ketosis, the ketone body concentration
in blood may be as high as 10-20 mmol/L. The ketone bodies are normally present in the daily urine in
trace amounts, while in pathology, 1 to 10 g (or even more) of ketone bodies per day is excreted in the
urine. Most commonly, ketonemia and ketonuria are observed in diabetes mellitus (the manifest keto-
sis symptoms are dependent on the extent of diabetes mellitus), as well as in prolonged starvation or in
steroid diabetes.

LECTURE 17

Subject: CATABOLISM OF SIMPLE PROTEINS

The biological value of proteins of animal and vegetable origin is determined by the constituent
amino acids, especially the essential ones. If food proteins contain all of the essential amino acids such
proteins are referred to as complete proteins. Any other food proteins are referred to as incomplete (or
partial) proteins. Plant proteins, as distinct from animal proteins, are, as a rule, less complete. The
hens egg protein has been recognized as an appropriate reference protein optimally corresponding to
physiological requirements of the organism. All food proteins should be compared for the amino acid
composition to the reference protein. The daily dietary protein intake for the adult human should
amount to 80-100 g, of which a half must be of animal origin.
Digestive mechanism for proteins
Proteolytic enzymes involved in the digestion of proteins and peptides are synthetized and secre-
ted in the digestive tract cavity as proenzymes, or zymogens. Zymogens are inactive and incapable of
digesting the intrinsic cell proteins. Proteolytic enzymes are activated in the intestinal lumen where
they act on dietary proteins.
The human gastric juice contains two structurally related proteolytic enzymes, pepsin and gastric-
sin, which are derived from a common precursor.
Pepsin is produced as a proenzyme, pepsinogen, in the major cells in the gastric mucosa. The
activity of pepsinogen is elicited either by hydrochloric acid (HCl) excreted from the cover cells of
stomach or autocatalytically, i.e. by the pepsin molecules themselves. The molecular mass of pepsino-
gen is 40,000. The pepsinogenic peptide chain contains pepsin (molecular mass 34,000), and a residual
polypeptide-inhibitor. Pepsin belongs to carboxyproteinases containing dicarboxy amino acid residues
in the active center, at pH optimum 1,5-2,5.
Proteins, both native and denatured, serve as substrates for pepsin. Denatured proteins are more
easily hydrolyzed. Food proteins are denatured by cooking or by treating with hydrochloric acid. The
following biological functions of hydrochloric acid are to be noted:
- activation of pepsinogen,
- provision for optimal pH in hydrolysis by pepsin and gastricsin juice,
- denaturation of food proteins,
- antimicrobial action.
The intrinsic proteins of stomach wall are protected against the denaturing action of hydrochloric
acid and digestive activity of pepsin by glycoprotein-containing mucous secretum. Pepsin, which is an
endopeptidase, exerts a fast cleavage of protein endo-peptide bonds formed by the carboxyl groups of
aromatic amino acids (phenylalanine, tyrosine, and tryptophan).
Gastricsin is close to pepsin by molecular mass (31,500). The optimal pH for gastricsin is about
3,5. Gastricsin hydrolyzes the peptide bonds formed by dicarboxylic acids. The pepsin/gastricsin ratio
in the gastric juice is 4:1. In patients with peptic ulcer, this ratio alters in favour of gastricsin.
Pepsin and gastricsin hydrolyze proteins to a mixture of polypeptides (also called peptones or
albumoses). The extent of protein digestion in the stomach is dependent on the food residence time
therein. Usually, this time is rather short (from 3 to 6 hours, depending on the digestive regime), there-
fore, the main portion of proteins is degraded in the intestine.
79
Proteolytic enzymes in intestine. The proteolytic enzymes are excreted to the intestine from the
pancreas as proenzymes: trypsinogen, chymotrypsinogen, procarboxypeptidases A and B, and proelas-
tase. The activation of these enzymes is effected via partial proteolysis of their polypeptide chains, i.e.
by cleaving off the fragment that acts as a shield for the active center of enzyme. The key process for
activating all of the proenzymes is the formation of trypsin. Trypsinogen secreted by the pancreas is
activated by the intestinal enterokinase, or enteropeptidase. In addition, the generated trypsin favours
the autocatalytic conversion of trypsinogen to trypsin. The mechanism for activation of trypsinogen
consists in the hydrolysis of one peptide bond resulting in the release of N-terminal hexapeptide as an
inhibitor for trypsin. Further, trypsin splits peptide bonds in other proenzymes to form active enzymes.
In such a way, three varieties of chymotrypsin, carboxypeptidases A, B, and elastase, are formed.
The intestinal proteinases hydrolyze the peptide bonds of dietary proteins and polypeptides pro-
duced by the gastric enzymes, to yield free amino acids. The endopeptidases (trypsin and chymotryp-
sin) and elastase facilitate the cleavage of the endopeptide bonds and reduce proteins and polypeptides
to smaller fragments. Trypsin mainly attacks the peptide bonds formed by the carboxyl groups of lysi-
ne and arginine; vis-a-vis the isoleucine peptide bonds, it is less active.
Chymotripsins are the most active towards the peptide bonds involving tyrosine, phenylalanine,
and tryptophan. The specificity of hydrolytic action of chymotrypsin is similar to that of pepsin.
Elastase hydrolyzed the bonds formed by aliphatic amino acids and imino acids, proline and hy-
droxyproline.
Carboxypeptidase A is a zinc-containing enzyme. It cleaves C-terminal aromatic and aliphatic ami-
no acids from polypeptides. Carboxypeptidase B splits C-terminal lysine and arginine residues only.
In the intestinal mucosa, dipeptidases occur that are capable of hydrolyzing dipeptides to their
two constituent amino acids. The dipeptidases are activated by cobalt and manganese ions and by cys-
teine.
The available variety of proteolytic enzymes provide for complete degradation of proteins to free
amino acids even in the contingency when the proteins remain unaffected by the action of gastric pep-
sin. Due to this happy circumstance, the patients that have been operated for a partial or complete ex-
cision of stomach retain the capability to assimilate dietary proteins.
Absorption of protein hydrolyzates
Amino acids are the major products of protein hydrolysis. Their intestinal uptake, as well as the
transport across other cell membranes, occurs via special amino acid transport systems. The amino
acid transport is an active transport and requires an appropriate Na
+
ion gradient, which is produced by
Na
+
, K
+
-ATPase of the intestinal epithelium membrane. Amino acids are absorbed in the intestine by
the secondary active transport. This is evidenced by the fact that the glycoside ouabain, an inhibitor
for Na
+
,K
+
-ATPase, also exerts a retarding effect on the amino acid transport.
There are known at least five amino acid carrier systems that specialize in the transport of:
1. neutral aliphatic amino acids,
2. cyclic amino acids,
3. basic amino acids,
4. acidic amino acids,
5. proline and hydroxyproline.
The amino acids of these groups complete for binding with a carrier of the corresponding tran-
sport system. In the transport of amino acids across the intestinal epithelium membrane Na
+
ions oc-
curs via a special carrier system. The sodium ions are pumped out of the cell by Na
+
,K
+
-ATPase, whi-
le the amino acids remain inside the cell.
There is also an alternative mechanism for the transport of amino acids across the cell membrane
of intestinal epithelium and of other cells; it is the so-called -glutamyl cycle. The amino acid transfer
is accomplished through the agency of a special enzyme, -glutamyl transferase, which is found wit-
hin the membrane of intestinal epithelium and other cells. The cofactor for this enzyme is a tripeptide,
glutathione, which is sufficiently abundant in the cell. The energy of glutathione peptide bonds is used
for the transfer of amino acid from the environment into the cell. The wide occurrence of the major en-
zymes of this transport cycle in the tissues is indicative of the enzymes importance for the transport of
amino acids in a large number of cells. In the intestine, the uptake of small amounts of dipeptides and
nonhydrolyzed proteins is possible. These are absorbed by endocytosis, and inside the cell they are
hydrolyzed by lysosomal proteinases.
80
The absorbed amino acids are delivered to the portal vein, then to the liver and, in a dissolved
state, are supplied in the blood to the tissues and organs. Amino acids are most actively consumed by
liver and kidney and, to a lesser extent, by other organs, especially brain.
Regulation of digestion
Digestion of food components is controlled by a system of hormone-like compounds formed in
the cells of the digestive tract: histamine, gastrin, secretin, cholecystokinin, etc. The secretion of di-
gestion regulators is elicited by the ingested food and is determined by its composition. On delivery of
food into the stomach histamine and gastrin are secreted which, in turn, provide for the secretion of
hydrochloric acid and pepsin involved in the digestion of proteins. The transit of stomach contents into
the duodenum stimulates the secretion of enterogastrone which, when excreted in the blood, inhibits
the gastric juice secretion.


Action of the major digestive hormones
The delivery of food into the intestine stimulates the secretion of a number of regulators (secretin,
cholecystokinin, pancreosimine, chymodenin, and enterocrinin) which, in turn, provide for a fast sec-
retion of pancreatic and intestinal juices for food digestion. Disturbances in secretory regulation cause
a disharmony of the digestive processes.

Pathology of digestion and absorption
Digestive disturbances are produced by a deficiency in enzymes and cofactors as well as by
biochemical malfunction of absorption in the intestine. A reduced secretion of hydrochloric acid and
pepsin (the so-called subacid gastritis or hypoaciditas), and, occasionally, a complete cessation (such
a state is referred to as achylia) do not affect significantly the overall digestibility of dietary protein.
This functional deficiency is a compensated for by an essential set of proteolytic enzymes under the
intestinal digestion conditions. However, a failure of hydrochloric acid production leads to the deve-
lopment of microbial flora and putrefactive processes in the stomach.
A shortage of proteolytic pancreatic enzymes (congenital or produced by mechanical obstruction)
leads to the discharge of undigested proteins with feces and, as a consequence, to a relative protein de-
privation. The indigested proteins are subject to the digestion by microorganisms of large intestine.
This process is called intestinal putrefaction of proteins. The protein putrefaction is concomitant with
the production of toxic materials: ammonia (NH
3
), hydrogen sulphide (H
2
S), amines (putrescine, cada-
verine), and alcohols (phenol, cresol, scatole, and indole). These products may exert a deleterious ef-
fect on the organism. Some of the noxious products are neutralized in the intestine, and others are ren-
dered harmless during absorption, mostly in the liver. Amines are inactivated by diaminooxidase
(DAO) type. The major route to detoxication of ammonia is urea synthesis in the liver. Indole, scatole,
phenol, and cresol are inactivated by conjugation with UDP-glucuronic acid and PAPS (3-phospho-
adenosine-5-phosphosulphate).

LECTURE 18

Subject: AMINO ACID CATABOLIC PATHWAYS IN TISSUES
81

The first step of protein renewal is proteolysis which effected by tissue proteinases, or cathepsins.
Cathepsins are located chiefly in the lysosomes, as many other hydrolytic enzymes. However, cathep-
sins also occur in other cell organelles: mitochondria, endoplasmic reticulum, and hyaloplasm. The ly-
sosomal cathepsins are most active in an acidic medium therefore they are referred to as acidic cathep-
sins. In the cell cytoplasm and other organelles, cathepsins are optimally active in neutral and weakly
basic media.
Proteolysis, as it is effected in tissues, is important for the renewal of proteins, for the disposal of
defective protein molecules, and for the energetic mobilization of endogenic proteins (especially in
starvation). Therefore, cathepsins perform not only a destructive function, but are also involved in re-
constructive processes. Cathepsins are capable of a limited proteolysis, i.e. they are apt to split off a
particular fragment from a polypeptide chain (this phenomenon is reminescent of the production of
enzymes from digestive proenzymes). The limited proteolysis liberates neuropeptides that perform
mediatory and hormonal functions. Prohormones produced by the endocrine glands are converted to
active protein hormones by the same mechanism.
Amino acid catabolic pathways. The catabolic pathways by which amino acids are degraded to
the end products may be divided into three groups:
1. catabolic pathways involving the conversion of NH
2
groups (any amino acid has at least one
o-NH
2
group),
2. pathways for the breakdown of amino acid carbon frameworks,
3. decarboxylation of amino acid o-COOH groups.
The third pathway is a version for the amino acid carbon framework conversion. It is employed in
the production of biogenic amines and will be dealt with later in discussing the processes involving
formation and degradation of mediators.
Conversion of o-amino groups of amino acids
In the organism tissue, the cleavage of an amino group from an amino acid leads to the produc-
tion of ammonia. This process is called deamination. Four types of deamination are known in nature:
reductive, hydrolytical, intramolecular, and oxidative. Oxidative deamination can proceed via a direct
route or an indirect route (transdeamination) in the human organism.
Direct oxidative deamination is effected for the glutamic acid:

COOH COOH COOH
| | |
H
2
N-CH Glutamate DH C=NH +H
2
O C=O
| | | + NH
3

(CH
2
)
2
NADNADH
2
(CH
2
)
2
(CH
2
)
2

| | |
COOH COOH COOH

glutamic acid iminoglutaric acid o-ketoglutaric acid

Transdeamination is the major route to amino acid deamination. It is accomplished in two steps.
The transamination reaction is a reversible one; it is catalyzed by enzymes aminotransferases, or tran-
saminases.

CH
3
COOH CH
3
COOH
| | | |
HC-NH
2
+ (CH
2
)
2
ALT C=O + (CH
2
)
2

| | | |
COOH C=O PP (B
6
) COOH HC-NH
2

| |
COOH COOH

alanine o-ketoglutaric acid pyruvic acid glutamic acid


COOH COOH COOH COOH
| | | |
HC-NH
2
+ (CH
2
)
2
AST C=O + (CH
2
)
2

| | | |
82
CH
2
C=O PP (B
6
) CH
2
HC-NH
2

| | | |
COOH COOH COOH COOH

aspartic acid o-ketoglutaric acid oxaloacetate glutamic acid

An aminotransferase is composed of an apoenzyme and coenzyme. Derivatives of pyridoxine (vi-
tamin B
6
), pyridoxal 5-phosphate (PP) and pyridoxamine 5-phosphate are coenzymes for aminotrans-
ferases. Amino transferases, alanine aminotransferase (ALT) and aspartate aminotransferase (AST),
have clinico-diagnostical value. They are organospecific enzymes (ALT-liver, AST-heart).
I ndirect oxidative deamination is effected for the other amino acids. This deamination is accom-
plished in two steps: transamination and direct deamination of glutamic acid.
Biosynthesis of nonessential amino acids
In the mammalian tissues, biosynthesis of only nonessential amino acids is possible; the essential
amino acids must be supplied in food. The starting materials for the biosynthesis of nonessential ami-
no acids are intermediates of carbohydrate catabolism, the Krebs cycle metabolites, and essential ami-
no acids. For example, alanine is produced from pyruvate via two routes:
1. transamination with the participation of alanine aminotransferase (ALT):

Pyruvate + glutamate ALT alanine + o-ketoglutarate

2. reductive amination involving alanine dehydrogenase (ADH):

Pyruvate + NH
3
+ NADH
+
+ H
+
+ H
2
O ADH alanine + NAD
+
.

Decarboxylation of amino acid o-COOH groups
Production of histamine. Histamine is synthetized from histidine by the action of histidine decar-
boxylase:

Figure 49. Conversion of histidine to histamine by histidine decarboxylase

Nearly all the tissues and organs contain histamine. Histamine is released from the mast cells by
the agents called histamine liberators. In large amounts histamine is produced in the gastric mucosa,
where it affects the secretion of pepsin and hydrochloric acid. Histamine in small amounts is always
present in the blood plasma and other biological fluids. Histamine serves as a mediator of allergic
reactions and is released in large amounts in pathologic processes.
Production of serotonin. Serotonin is derived from tryptophan.
Serotonin acts as a mediator in the nervous system and as a local functional regulator for the peri-
pheral organs and tissues.
Production of -aminobutiric acid (GABA). GABA is produced from glutamic acid by the action
of glutamate decarboxylase:

HOOC-CH
2
-CH
2
-CH-COOH GDC HOOC-CH
2
-CH
2
-CH
2
-NH
2
+ CO
2

|
NH
2
PP (B
6
)
glutamate GABA

The synthesis proceeds in the inhibitory synapses of the nervous system; for these, GABA acts as
a mediator.

83

Figure 50. Serotonin production

I nactivation of biogenic amines. A route to detoxication of biogenic amines is the oxidative dea-
mination with the involvement of aminooxidases: monoaminoaxidase (MAO) type and diaminooxidase
(DAO) type.

R-CH
2
-NH
2
+ O
2
+ H
2
O Aminooxidase R-HC=O + NH
3
+ H
2
O
2

biogenic amine aldehyde

Aldehydes produced by deamination of biogenic amines are oxidized to organic acids by alde-
hyde dehydrogenases:

R-HC=O + NADH
+
+H
+
+ H
2
O R-COOH + NAD
+


LECTURE 19

Subject: ROUTES TO METABOLIC DETOXIFICATION OF AMMONIA

In the organism, ammonia (NH
3
) is produced by the following processes deamination of:
1. amino acids,
2. biogenic amines (histamine, serotonin, etc.),
3. purine bases (adenine and guanine),
4. pyrimidine bases (uracil, thymine, cytosine),
5. amino acid amides (asparagine and glutamine).
Ammonia is a very toxic compound, especially for nervous cells. Its accumulation in the orga-
nism elicits excitation of the nervous system.

84
Mechanism of toxication of ammonia

COOH COOH COOH
| | |
C=O + NH
3
C=NH GDH HC-NH
2

| -H
2
O | |
(CH
2
)
2
(CH
2
)
2
NADH
2
NAD (CH
2
)
2

| | |
COOH COOH COOH

o-ketoglutaric acid iminoglutaric acid glutamic acid

NH
3
is inhibitor of Krebs cycle. For this reason, to counteract the adverse effects, mechanisms for
neutralization of ammonia are operative in the organism tissues. These mechanisms include:
1. production of urea;
2. reductive amination, or transreamination;
3. production of amino acid amides (asparagine and glutamine);
4. production of ammonium salts.
The major route to detoxification of ammonia is urea synthesis. In the liver urea was formed from
ammonia and carbonic acid. The urea cycle also refer to as the arginine-urea cycle, or ornithine cycle.

Urea production cycle (ornithine cycle)


Figure 51. Scheme of urea production: 1 L-ornithine; 2 carbamoyl phosphate; 3 L-citrulline; 4
argininosuccinate; 5 fumarate; 6 L-arginine; 7 urea; L-Asp: L-aspartate; CPS-1: carbamoyl phosphate
synthetase I; OTC: Ornithine transcarbamylase; ASS: argininosuccinate synthetase; ASL: argininosuccinate
lyase; ARG1: arginase 1.

85
To produce one urea molecule, three ATP molecules are needed to be consumed. Urea is a
material which is inert towards the living organism. The major site for its formation in the organism is
the liver which contains all the enzymes involved in urea production.
Disturbances in the hepatic function lead to a decreased urea production, with the resultant dimi-
nution of urea blood concentration and urea excretion in the urine.
Reductive amination. Viewed from the standpoint of an ammonia-binding process, it is little ef-
fective, since it requires a significant amount of o-ketoglutarate for its effectuation.

COOH COOH COOH
| | |
C=O + NH
3
C=NH GDH HC-NH
2

| -H
2
O | |
(CH
2
)
2
(CH
2
)
2
NADH
2
NAD (CH
2
)
2

| | |
COOH COOH COOH
o-ketoglutaric acid iminoglutaric acid glutamic acid

Production of asparagine and glutamine. It is important a subsidiary route to binding ammonia.
It proceeds with the involvement of asparagine synthetase and glutamine synthetase:

COOH COOH
| Asparagine |
HC-NH
2
+ NH
3
synthetase HC-NH
2
+ H
2
O
| |
CH
2
ATPADP CH
2

| |
COOH CONH
2

aspartic acid asparagine

COOH COOH
| Glutamine |
HC-NH
2
+ NH
3
synthetase HC-NH
2
+ H
2
O
| |
(CH
2
)
2
ATPADP (CH
2
)
2

| |
COOH CONH
2

glutamic acid glutamine

This process is active in nervous and muscle tissues and in kidney.
Ammonium salt production. Glutamine and, to a lesser extent, asparagine are believed to be a
kind of vehicles for ammonia, since two species, after their formation in the tissues, are delivered in
the bloodstream to the kidneys to be hydrolyzed by the action of specific enzymes, glutaminase and
aspartaginase:
COOH COOH
| |
HC-NH
2
+ H
2
O Glutaminase HC-NH
2
+ NH
3

| |
(CH
2
)
2
(CH
2
)
2

| |
CONH
2
COOH
glutamine glutamic acid

Ammonia, liberated in the tubules of the kidney, is neutralized to ammonium salts:

NH
3
+ H
+
+ Cl
-
NH
4
Cl, which are excreted in the urine.

Amino acids as medicinal preparations. In the practical medicine, preparations on the basis of
protein hydrolyzates and individual amino acids are used. Methionine and other methionine-rich hy-
drolyzates are used as lipotropic factors, and in treating protein deficiency in chronic diseases. For cli-
86
nical purposes, glutamic and aspartic acids are widely used, which play a decisive role in the detoxifi-
cation of ammonia and in other synthetic reactions of amino acid metabolism.

LECTURE 20

Subject: CONVERTION OF AMINO ACIDS TO SPECIALIZED PRODUCTS

Amino acids are widely used for the biosynthesis of a large variety of nonproteinic nitrogenous
materials: choline, phosphatides, creatine, mediators (including biogenic amines), pigments, vitamins,
coenzymes, porphyrins, purine and pyrimidine bases, etc.
Production of taurine. Taurin is a biogenic amine; it is produced from cysteine:

H
2
C-SH H
2
C-SO
2
H H
2
C-SO
3
H H
2
C-SO
3
H
| | | |
H-C-NH
2
H-C-NH
2
H-C-NH
2
CH
2

| | | |
COOH COOH COOH NH
2
cysteine cysteine sulfinic cysteic taurine
acid acid
This compound is synthetized in various organs. In the liver, taurine is involved in conjugation
reactions with bile acids. In the nervous system, taurine is presumably carable of acting as a synaptic
mediator.
Biosynthesis and breakdown of creatine. In the human and animal tissues, creatine synthesis
proceeds with the involvement of three amino acids: arginine, glycine, and methionine. Creatine syn-
thesis proceeds in two steps. The first step involves the formation of guanidinoacetic acid from argini-
ne and glycine with the participation of the enzyme glycine-amidine transferase:

NH
2
NH
2

| |
C=NH C=NH NH
2

| NH
2
| |
NH | NH (CH
2
)
3

| + CH2 | + |
(CH
2
)
3
| CH
2
H-C-NH
2

| COOH | |
H-C-NH
2
COOH COOH
|
COOH guanidinoacetic

L-arginine glycine acid L-ornihine
The first step proceeds actively in the kidney and pancreas. At the second step, methylation of
guanidinoacetic acid through the assistance of guanidinoacetate methyltransferase takes place. The
active form of methionine, S-adenosylmethionine, acts as a methyl group donor:

NH
2
NH
2

| |
C=NH C=NH
| S-adenosylmethionine |
NH | N-CH
3

| \ S-adenosylhomocysteine |
CH
2
CH
2

| |
COOH COOH
guanidinoacetic acid creatine

This step takes place in the liver and the pancreas which provide for the conditions and for the
appropriate enzyme required for the synthesis of creatine from guanidinoacetic acid. It is commonly
believed that the creatine that has been synthesized in the liver and in the pancreas is supplied via
blood circulation to other organs and tissues: brain, skeletal muscles, heart, etc. In the cells, creatine is
87
involved in the energy transfer via reversible transphosphorylation with ATP. A product of creatine
breakdown is creatinine produced via a nonenzymic pathway:
_________
| |
H
2
N-C-N-CH
2
-COOH HN-C-N-CH
2
-C=O
|| | -H
2
O || |
HN CH
3
HN CH
3
creatine creatinine

About 2% of creatine contained in the organism is liable to conversion to creatinine. In small
amounts, creatine and creatinine are present in the blood plasma. In the creatine-synthetising organs
(kidney, liver, pancreas), creatine concentrations are small (0.1-0.4 g/kg). In highest amounts, creatine
is found in skeletal muscles (25-55 g/kg), heart (15-30 g/kg), and brain tissue (10-15 g/kg).
Urinary creatine is found only in children. In adult humans, creatinine is excreted in the urine (4.4
to 17.6 mmol per day); creatinine excretion has been found to be directly related to the muscular deve-
lopment of the human body. Creatine found in the urine of an adult human is indicative of pathology.
Production of certain vitamins and coenzymes from amino acids. In human tissues, nicotinami-
de (vitamin PP) is produced from tryptophan. Therefore, tryptophan supplied in food can in part com-
pensate for a deficit of dietary nicotinamide. Cysteine is used for the biosynthesis of coenzymes of
pantothenic acid (vitamin B5), 4-phosphapantetheine, and HS-coA. Glutathione, which acts as a coen-
zyme for certain oxidoreductases and transferases (glutathione reductase, glutathione peroxidase, glu-
tathione-S-transferase, -glutamyltranspeptidase, etc.), is produced from cysteine, glutamic acid, and
glycine by a two-step reaction. At the first step of this synthesis, glutaminylcysteine synthetase is ope-
rative, and at the second step, glutathione synthetase.
Production and breakdown of mediators. Mediators are produced in the nervous tissue and cer-
tain other cells. Neuromediators, which are generated in the nerve endings, are transmitter substances
that participate in the transmission of the nervous impulse to other nervous cells or peripheral organs
and tissues. Tissue mediators take part in the metabolic tissue regulation.
In the production of a number of mediators, of special importance is the reaction of amino acid
decarboxylation specifically catalyzed by decarboxylases (enzymes that contain pyridoxal phosphate
as a coenzyme):

R-CH-COOH decarboxylase R-CH
2
-NH
2
+ CO
2

|
NH
2
PALP biogenic amine
amino acid

The decarboxylation products are amines exhibiting a high biological activity. For this reason,
they are referred to as biogenic amines. Most mediators belong to this group of compounds.

involvement of aminooxidases:
aminooxidase
R-CH
2
-NH
2
+ O
2
+ H
2
O R-C=O + NH
3
+ H
2
O
2

\H

Aminooxidases are differentiated into a monoaminooxidase (MAO) type and a diaminooxidase
(DAO) type. FAD serves as a coenzyme for MAO, and pyridoxal phosphate, for DAO (the reaction
requires the presence of Cu
2+
ions). MAO is bound with the cell mitochondria, and DAO is found in
the cytoplasm. In small amounts these enzymes are present in the blood. Primary, secondary, and ter-
tiary amines are inactivated by MAO, while histamine, putrescine, cadaverine, and to a lesser extent,
aliphatic amines are mostly inactivated by DAO. Aldehydes produced by deamination of biogenic
amines are oxidized to organic acids by aldehyde dehydrogenases:

R-C=O + NADH
+
+ H
+
+ H
2
O R-COOH + NAD
+

\H

Production of catecholamines. Catecholamines constitute a group of biogenic amines with me-
diatory and hormonal functions. They are produced from phenylalanine and tyrosine. The following
biogenic amines are derived from phenylalanine and tyrosine: phenylethylamine, phenylethanolamine,
88
3,4-dihydroxyphenylalanine (DOPA), noradrenalin, adrenalin, dopamine (3,4-dihydroxyphenethylami-
ne).


Figure 52. Scheme of adrenalin and noradrenalin production

The catecholamines that exhibit mediatory and hormonal functions to a great extent are adrenalin
and noradrenalin. Noradrenalin and adrenalin are synthesized in the chromaffin cells of substantia me-
dullaris of the adrenal glands (for performing a hormonal function), in the adrenergic synapses of the
brain, and in the sympathetic nerve endings of the vegetative nervous system.
Adrenalin and noradrenalin are inactivated via two pathways:
- involvement of monoaminooxidase, via deamination,
- involvement of catechol O-methyltransferase, via methylation.
Glycine. Metabolites and pharmaceuticals excreted as water-soluble glycine conjugates include
glycocholic acid and hippuric acid formed from the food additive benzoate. Many drugs, drug metabo-
lites, and other compounds with carboxyl groups are excreted in the urine as glycine conjugates. Gly-
cine is incorporated into creatine, the nitrogen and o-carbon of glycine are incorporated into the pyrro-
le rings and the methylene bridge carbons of heme, and the entire glycine molecule becomes atoms
4, 5, and 7 of purines.
Cysteine. L-Cysteine is a precursor of the thioethanolamine portion of coenzyme A and of the
taurine that conjugates with bile acids such as taurocholic acid.
Methionine. S-Adenosylmethionine, the principal source of methyl groups in the body, also con-
tributes its carbon skeleton for the biosynthesis of the 3-diaminopropane portions of the polyamines
89
spermine and spermidine. It is donor of methyl groups for synthesis of adrenalin, thymine, choline,
acetylcholine, phosphatidylcholine, etc.
Histidine. Decarboxylation of histidine to histamine is catalyzed by a broad-specifity aromatic L-
amino acid decarboxylase. Histidine compounds present in the human body include ergothioneine,
carnosine, and dietary anserine. Urinary levels of 3-methylhistidine are unusually low in patients with
Wilsons disease.
Ornithine, and arginine. Arginine is the formamidine donor for creatine synthesis and via orni-
thine to putrescine, spermine, and spermidine. Arginine is also the precursor of the intercellular signa-
ling molecule nitric oxide (NO) that serves as a neurotransmitter, smooth muscle relaxant, and vasodi-
lator. Synthesis of NO, catalyzed by NO synthase, involves the NADPH-dependent reaction of L-argi-
nine with O
2
to yield L-citrulline and NO.
Tryptophan. Following hydroxylation of tryptophan to 5-hydroxytryptophan by liver tyrosine hy-
droxylase, subsequent decarboxylation forms serotonin (5-hydroxytryptamine), a potent vasoconstric-
tor and stimulator of smooth muscle contraction. Catabolism of serotonin is initiated by monoamine
oxidase catalyzed oxidative deamination to 5-hydroxyindoleacetate. The psychic stimulation that fol-
lows administration of iproniazid results from its ability to prolong the action of serotonin by inhibi-
ting monoamine oxidase. In carcinoid (argentaffinoma), tumor cells overproduce serotonin. Urinary
metabolites of serotonin in patients with carcinoid include N-acetylserotonin glucuronide and the gly-
cine conjugate of 5-hydroxyindoleacetate.
Kidney tissue, liver tissue, and fecal bacteria all convert tryptophan to tryptamine, then to indole
3-acetate. The principal normal urinary catabolites of tryptophan are 5-hydroxyindoleacetate and indo-
le 3-acetate.
Tyrosine. Neutral cells convert tyrosine to epinephrine and norepinephrine. While DOPA is also
an intermediate in the formation of melanin, different enzymes hydroxylate tyrosine in melanocytes.
DOPA decarboxylase, a pyridoxal phosphate-dependent enzyme, forms dopamine. Tyrosine is also a
precursor of triiodothyronine (T
3
) and thyroxine (T
4
).
Aspartic and glutamic amino acids. Ammonia may be toxic to the brain in part because it reacts
with o-ketoglutarate to form glutamate. The resulting depleted level of o-ketoglutarate then impairs
function of the tricarboxylic acid (TCA) cycle, or the Krebs cycle in neurons. Formation of glutamate
is catalyzed by mitochondrial glutamine synthase. One function of glutamine is to sequester ammonia
in a nontoxic form. Also production of asparagine is catalyzed by asparagine synthase. In kidney tis-
sue glutamine is donor of ammonia for production of ammonium salts, which excreted with urine.
Glutamine and asparagine are donors of ammonia for urea synthesis in liver. Aspartate is needed for
urea production. Glutamate and aspartate participate in synthesis of purines, pyrimidins, and malate-
aspartate shuttle mechanism. GABA is a product of glutamate decarboxylation. L-glutamate dehydro-
genase occupies a central position in nitrogen metabolism.
Amino acid metabolism control in the organism. Amino acid balance in the human organism is
dependent on the nutritive value of dietary proteins. Lack of any of the essential amino acids in the ali-
mentary diet or a prolonged deficit of half-essential amino-acids entails a disorder in the utilization of
other amino acids (both essential and nonessential) in protein biosynthesis. Amino acids transport to
the cells of different organs is apparently determined by the efficiency of the carrier systems involved
with respect to different groups of amino acids. In the kidneys the reabsorption of amino acids from
the urine into the blood occurs; owing to this recovery mechanism, only a small amount of amino
acids is lost into the urine.
The dietary supply of amino acids is one of the factors that regulates amino acid metabolism. An
excessive consumption of food high in proteins results in an increased delivery of amino acids to the
liver. The amino acids supplied favour an increased activity of the liver enzymes in the catabolic con-
version of amino acids to the end products. This mechanism provides for the removal of an amino acid
excess supplied to the organism. During starvation, on the contrary, an active breakdown of tissue pro-
teins to free amino acids takes place.
The amino acid balance control at various stages of amino acid transport can be effected by
means of the intestinal epithelium (in absorption), peripheral tissues (in intracellular penetration), and
the tubules of the kidney (in reabsorption). Transmembrane amino acid transport is stimulated chiefly
by insulin. Under physiological conditions, insulin, somatotropin, thyroid hormones, and male and fe-
male sex hormones facilitate the utilization of amino acids in protein biosynthesis and other synthetic
processes.
90
Since amino acids constitute the major source of nitrogen for all the nitrogenous materials, they
are responsible for the nitrogen balance in the organism. Nitrogen balance is the difference between
the amount of nitrogen supplied to the organism in nitrogen-containing nutrients (chiefly proteins and
amino acids) and that of nitrogen-containing materials discharged from the organism in the urine, fe-
ces, and sweat. If the amount of nitrogen supplied is equal to that lost from the organism, a nitrogen
equilibrium sets in. If the amount of nitrogen ingested is larger than that excreted from the organism,
the nitrogen balance is said to be positive. With the nitrogen eliminated superior to that supplied, the
nitrogen balance is negative. The positive nitrogen balance is observed during the active synthesis of
proteins from amino acids (during prenatal development of the fetus, during juvenile growth, in the
diet rich in meat, in the administration of anabolic hormones, etc.). The negative nitrogen balance
develops under complete protein deprivation, under prolonged reduction of motor activity, in grave
chronic diseases, burns, and other states adverse to the normal protein digestion and amino acid absor-
ption, or conducive to the degradation of tissue protein.

LECTURE 21

Subject: INTERRELATION OF MAJOR METABOLIC ROUTES IN THE
ORGANISM. METABOLISM OF NUCLEOPROTEINS

I nterrelation of metabolic routes in the human organism. The metabolisms of the monomers
basically important for the living systems: amino acids, monosaccharides (glucose), fatty acids, and
mononucleotides, are intimately interrelated. This all-pervading interdependence effected through the
so-called key metabolites that intervene as common links in the catabolic and anabolic pathways of the
monomers. Pyruvate, acetyl~ScoA, o-glycerol phosphate, and the Krebs cycle intermediates (oxaloa-
cetate, malate, fumarate, succinyl~ScoA, o-ketoglutarate, isocitrate, and citrate) can be cited among
these key metabolites. Pyruvate is a crossing point for the routes of synthesis and breakdown of gluco-
se, other monosaccharides, and some of the amino acids. Acetyl~ScoA is a more branched site for me-
tabolic pathways. Acetyl~ScoA bridges the gaps between monosaccharides, amino acids, and lipids,
i.e. opens up possibilities for conversions of amino acids (finally, proteins) and glucose (in a wider
sense, carbohydrates) to lipids. o-Glycerol phosphate serves as an accessory link between carbohydra-
tes and lipids. It participates in the conversion of carbohydrates to certain lipids (triglycerides and
phosphoglycerides) and, vice versa, of glycerol-containing lipids to carbohydrates.
Ample opportunities for the interconversion of monomers are provided by the intermediary pro-
ducts of the Krebs cycle. The Krebs cycle is a switching centre for all major catabolic and anabolic
routes. For example, fatty acids, serving as a source for acetyl-ScoA, are convertible to carbohydrates
(gluconeogenesis proceeds via the Krebs cycle oxaloacetate), to amino acids (via oxaloacetate and o-
oxoglutarate to aspartic and glutamic acids) and to porphyrins (via succinyl-ScoA).
Since monomers are structural units for biopolymers, relative interchangeability of proteins, car-
bohydrates, and lipids as major nutritional components of the food, is possible. The acceptable limits
for this interchangeability provide the clue to an understanding both of an eventual metabolic disorder
as produced by monotonous dietary regimen and of the mechanism of metabolic compensation in star-
vation, i.e. when the essential materials are produced at the expense of the endogenic reserves of other
materials in the organism.

Metabolism of purine and pyrimidine mononucleotides
Mammals are not in need of dietary supply of nitrogenous bases or nucleotides despite the ability
of the mammalien organism to assimilate directly these nutrients with food rich in nucleuc acids. In
mammalian tissues the consumed purine and pyrimidine nucleotides are continually renewed via their
synthesis from simple compounds.
Nucleotide phosphates, which are present in normal cells in small and relatively constant amo-
unts, act as a link in the pathways of supply and consumption of mononucleotides:
Nucleic acids Synthesis of nucleic acids
Nucleotide Synthesis of nucleotide
Synthesis from phosphate coenzymes
simple compounds Breakdown to end products
91
Biosynthesis of purine mononucleotides. The starting compound in the synthesis of purine
nucleotides is phosphoribosyl pyrophosphate (PRPP), which can be derived from ribose-5-phosphate
and ATP. The prime purine mononucleotide that crowns a long chain of synthetic reactions is inosine
monophosphate (IMP), which is a parent species for generating other purine nucleotide phosphates via
AMP and xantosine monophosphate (XMP). The genesis of the purine mononucleotide (AMP):

IMP Adenylosuccinate AMP
IMP + aspartate Adenylosuccinate fumarate + AMP

Figure 53. Scheme of purine synthesis

Breakdown of purine mononucleotides. Hypoxanthine is a naturally occurring purine derivative.
It is also a deamination product of adenine. Xanthine is a product on the pathway of purine
degradation, oxidation of hypoxanthine by xanthine oxidase. Also, by action of xanthine oxidase on
xanthine forms the end product of purines degradation uric acid, which excretes with urine.


AMP Hypoxanthine Xanthine Uric acid

Biosynthesis of pyrimidine mononucleotides (AMP, GMP). The starting compounds in the syn-
thesis of pyrimidine mononucleotides are carbomoyl phosphate and aspartic acid. They lead, through a
long chain of reactions, to uridine 5-monophosphate (UMP) and other pyrimidine mononucleotides.
The constituent atoms of the pyrimidine ring are supplied by carbomoyl phosphate and aspartic acid.
Breakdown of pirimidine mononucleotides (TMP, UMP, CMP). The end products of pirimidine
nucleotides degradation in the tissues are NH
3
, CO
2
, H
2
O and |-alanine. Ammonia and CO
2
are used
for urea production in the liver. In the muscles |-alanine is used for production of anserin and carno-
sin. So, the end product of pirimidine nucleotides breakdown is urea. Urea and |-alanine are excreted
with urine.
Biosynthesis of deoxyribonucleotides, precursors to DNA, is effected by reducing ribose to 2-
deoxyribose within the preformed nucleotide. A special protein, thioredoxin, bearing two SH groups in
its molecule, acts as a reductant in this process.

LECTURE 22

Subject: HEMOPROTEIN METABOLISM

A specific feature of hemoproteins is the metabolic involvement of the nonprotein moiety of these
conjugated proteins. The hemoglobin of blood erythrocytes and of marrow cells accounts for a major
portion (about 83%) of hemoproteins in the human organism. The remainder is myoglobin of skeletal
muscles and heart (about 17%) and cellular hemoproteins cytochromes, catalase, and peroxidase (1%).

Synthesis of hemoproteins
Glycine and succinyl~ScoA are the starting compounds in heme synthesis. The reaction involving
the pyridoxal-assisted enzyme o-aminolevulinate synthetase yields o-aminolevulinic acid. Two mole-
92
cules of o-aminolevulenic acid are combined with the participation of porphobilinogen synthetase, to
form porphobilinogen, a direct precursor of porphyrins. One of these is coproporphyrin III which is
directly converted to protoporphyrin IX. The insertion of iron ions (Fe
2+
) into the protoporphyrin IX
ring is effected with the assistance of ferrochelatase. At the ultimate step, heme becomes complexed
with globin to form hemoglobin or myoglobin. In the synthesis of other hemoproteins, heme adds to
the specific protein moiety of cytochromes or other hemo-containing enzymes. It these is not a
sufficient quantity of protein to bind the reaction step of porphyrin synthesis.

COOH NH
2
COOH
| | |
CH
2
+ CH
2
o-aminolevulinate CH
2

| | synthetase |
CH
2
COOH C=O
| |
C=O CH
2

\ScoA |
NH
2
succinyl-ScoA glycine o-aminolevulinic acid

The enzymes involved in heme biosynthesis are found in the marrow, nucleated erythrocytes,
liver, kidneys, and intestinal mucosa. The reactions leading to o-aminolevulinic acid proceed in the
mitochondria; the production of porphobilinogen and the subsequent synthesis of coproporphyrinogen
III occur in the cytoplasm, and the synthesis of heme from coproporphyrinogen III, in mitochondria.



Figure 54. Scheme of heme

Breakdown of hemoproteins
The first stage of hemoprotein catabolism. In the human organism, about 9g of hemoproteins per
day is degraded, mainly owing to the breakdown of erythrocytic hemoglobin. Erythrocytes undergo
degradation (hemolysis) in the bloodstream or in the spleen in the range of about 120 days.
Hemoglobin, released from the erythrocytes in the blood, becomes bound with haptoglobin (a
group of glycoproteins in the o
2
-globulin fraction of blood plasma) and is supplied, as a hemoglobin-
haptoglobin complex, to the cells of the reticulo-endothelial system (RES), chiefly to those of the sple-
en. Hemoglobin is oxidized to methemoglobin (Fe
3+
) to be subsequently degraded. Haptoglobin is split
off to pass into the blood. In RES cells, the first stage of hemoglobin degradation takes place, leading
to bilirubin production. In the first step, heme is converted to biliverdin by the enzyme heme oxygena-
se (HOXG).
HMOX
heme biliverdin + Fe
3+

/ \
H
+
+ NADPH
+
NADP
+

O
2
CO

93
NADPH
+
is used as the reducing agent, molecular oxygen enters the reaction, carbon monoxide
CO is produced and the iron is released from the molecule as the ferric ion (Fe
3+
).
In the second reaction, biliverdin (biliverdin is a bile pigment of green colour) is converted to bili-
rubin by biliverdin reductase (BVR). Biliverdin can be reduced to bilirubin, a yellow-red pigment:

BVR
biliverdin bilirubin
/ \
H
+
+ NADPH
+
NADP
+



Bilirubin

Globin is hydrolyzed by the spleen cathepsins to amino acids. Bilirubin is sparingly soluble in
water and, when released into the blood, it becomes bound with the plasma protein albumin. The
albumin-bilirubin complex is the most important normal transport form for bile pigments. In the
bloodstream, bilirubin is carried over to the liver cells. Since bilirubin is a lipophil, it passes easily
across the liver cell membrane and, in doing so, liberates itself from albumin.
The second stage of hemoprotein conversion takes place inside the liver cells. Conjugated forms
of bilirubin, bilirubin glucuronides, are produced in the liver. UDP-glucuronic acid acts as a donor of
glucuronic acid.


Figure 55. Chemical structure of UDP-GA

Bilirubin is transported into the liver, where it is conjugated with glucuronic acid to become mo-
re water soluble. The reaction is catalyzed by the enzyme UDP-glucuronide transferase (UDP-GUTF)
glucuronosyltransferase to yield bilirubin monoglucuronide (20%) and bilirubin diglucuronide (80%).
Bilirubin glucuronides are compounds readily soluble in water.

UDP-GUTF
bilirubin + 2 UDP-glucuronate bilirubin diglucuronide
\
2 UMP + 2 Pi

Bilirubin glucuronides are capable, to a small extent through, of diffusing into the blood capillary.
For this reason, two bilirubin forms are present in the blood plasma: unconjugated (also referred to as
indirect, or free) bilirubin, and conjugated (direct or bound) bilirubin. The former one accounts for
about 75% of the total blood plasma bilirubin, and the latter, for about 25%.
The third stage of hemoprotein degradation. Bilirubin glucuronides are excreted in the intestinal
bile where the third, final stage of hemoprotein degradation occurs. In the bile tracts, glucuronic acid

94


Figure 56. Bilirubin diglucuronide

is split off bilirubin glucuronides, with the recovery of unconjugated bilirubin. In the small intestine, a
small portion of bilirubin can be absorbed and, through the portal vein, be supplied to the liver to be
again excreted in the bile from the intestine. This hepatoenteric circulation, bearing resemblance to
bile acid circulation, is presumably by the intestinal bacteria or by the reductases of intestinal mucosa.
In the small intestine, bilirubin is converted to mesobilirubin and then to mesobilinogen (or urobilino-
gen). The latter is absorbed in the small intestine and through the portal vein is delivered to the liver in
which urobilinogen is irreversibly degraded to mono- and dipyrroles. The intact urobilinogen is retur-
ned in the bile to the intestine. In the large intestine, mesobilirubinogen (urobilinogen) is reduced by
anaerobic bacteria to stercobilinogen which like urobilinogen, is a colourless substance. Most stercobi-
linogen is excreted in the feces and undergoes rapid oxidation with atmospheric oxygen to stercobilin,
an orange-yellow pigment, chiefly responsible for the colour of the feces.
A small amount of stercobilinogen is absorbed in the rectum, and through the hemorrhoidal vein
system, in bypass to the liver, is delivered in the bloodstream to the kidneys to be excreted in the urine.
Urinary stercobilinogen, like fecal stercobilinogen, is oxidized to stercobilin which is in part responsi-
ble for the light-yellow colour of the normal urine. It was formerly believed that urobilinogen was ex-
creted in the urine (hence the name coined for this material) to be oxidized to urobilin exhibiting the
same colour as stercobilin. Presumably, the resemblance in colour might have led to erroneous conclu-
sions, since urobilin and stercobilin structurally are different species. Normally, urobilinogen is excre-
ted neither in urine, nor in feces.
Thus, the intermediate product of heme breakdown, bilirubin, is normally not accumulated in the
blood, but is rapidly entrapped by the liver cells. The concentrations (absolute and relative) of diffe-
rent forms of bilirubin in the blood allow on the other hand, on the bilirubin conversion in the liver.
Normally, the total bilirubin concentration in the blood serum amounts to 8-20 mcmol/litre (here un-
conjugated bilirubin accounts for 75%).
In humans, the end product of bilirubin conversion, stercobilinogen, is chiefly excreted in the
feces (about 300 mg per day) and, to a lesser extent, in the urine (about 1-4 mg per day). The composi-
tion of the pigments excreted in the feces and urine is influenced by the intestinal microflora. In neo-
nates, whose intestine is devoid of microflora, bilirubin being excreted is oxidized to biliverdin, which
is green in colour; for this reason, the meconium (newborns feces) retains a dark green grass-like co-
lour. In infants, within the first year of life, as the intestine becomes populated with microflora, the
composition of excreted pigments and the colour of feces gradually change. The greenish fecal colour,
produced by the presence of biliverdin, disappears, since the intestinal bacteria become increasingly
active in the reduction of bilirubin to stercobilinogen.

Pathology of bile pigment metabolism
The production, conversion, and excretion of bilirubin in the organism may become disturbed
owing to a variety of factors. An increased concentration of bilirubin in the blood leads to its deposi-
tion in tissues, including skin and mucosa, and imparts to these a yellow-brownish colour (the color of
bilirubin). Such states of the organism are referred to as jaundices. A variety of jaundices are known,
of which typical are:
- hemolytic jaundice,
- parenchymatous (or hepatocellular) jaundice,
- obstructive jaundice.
95
Hemolytic jaundicemay arise from a number of causes conducive to an extensive intravascular
(or tissue) breakdown of erythrocytes, or to their tissue breakdown (in RES cells). A large amount of
unconjugated bilirubin, excreted from RES cells in the bloodstream, fail to become conjugated in the
liver, which results in a high bilirubin level in the blood. Owing to an excess of excreted stercobilin
and urobilin, the feces acquire an intense, nearly dark coloration, and the abundantly excreted urobilin
colours the urine intensely yellow-orange.
Parenchymatous jaundice rises becuse of impaired liver cells (as afflicted with viruses or toxic
hepatotropic compounds); the liver cells become more permeable to invading agents, including biliru-
bin glucuronides normally excreted from hepatic cells in the blood in small amounts. The impaired he-
patic cells exhibit a reduced activity towards the uptake of bilirubin from the blood and fail to provide
for the normal production of bilirubin glucuronides in them. For this reason, despite the normal hemo-
lysis, the concentrations of unconjugated and conjugated bilirubin increase, albeit to a lesser extent as
in hemolytic jaundice. Because of a diminished excretion of stercobilin and urobilin, the feces and the
urine are but slightly coloured. However, a small quantity of urinary unconjugated bilirubin, normally
absent in the urine, is observed.
Obstructive jaundice as its name implies arises due to an impeded outflow of bile in the intestine.
As a consequence, conjugated bilirubin is reexcreted from the liver cells in the blood. The blood con-
tains an increased amount of conjuated bilirubin which, being a readily soluble compound, is excreted
at a higher rate in the urine. Because of this, the urine acquires a beer-like colour, and is discharged
with yellow bright foam. The feces, devoid of bile pigments, are imparted a grey-white, clay-like co-
lour.
I nogenous jaundice (jaundice in newborns) is believed to be of physiological origin. This disease
arises from the developmental insufficiency of glucuronosyltransferase, the bilirubin conjugation en-
zyme. Therefore, an increased degradation of erythrocytes, irrespective of its origin, leads to an enhan-
ced level of unconjugated bilirubin in the blood and, consequently, to an icteric indisposition. Com-
monly, physiological jaundice in newborns is transient within two weeks as the hepatic secretion of
glucuronosyltransferase increases. In premature infants, physiological jaundice may be of longer dura-
tion. However, a prolonged increase of unconjugated bilirubin concentration in the blood may consti-
tute a health hazard due to the toxic effect of bilirubin on the developing brain. Occasionally, convul-
sions and irreversible nervous system disturbances are observed in infants. In adults, the cerebral cells
are little permeable to bilirubin and, as a rule, hyperbilirubinemia evokes no complications. In certain
instances, in order to stimulate the activity of glucuronosyltransferase, the administration of phenobar-
bital is recommendable, which increases the enzyme secretion and alleviates the icteric processes.
The impaired pigment metabolism is also observed in intestinal dysbacteriosis produced by sup-
pression of normal microflora in the intestine (for example, as a result of prolonged therapy with tetra-
cycline antibiotics). In dysbacteriosis, intermediary products of bilirubin reduction are excreted in the
feces; under deep suppression of the intestinal microflora, bilirubin itself, when oxidized to biliverdin,
colours the feces greenish.

LECTURE 23

Subject: NEUROENDOCRINE CONTROL OF METABOLISM

The endocrine system includes special glands whose cells function to secrete chemical regulators,
commonly referred to as hormones, into the internal media of organism, i.e. blood and lymph. The
notion hormone (from the Greek hormaion, to incite, to set in motion) was coined by Beyliss and
Starling in 1905. Currently, hormones are called compounds that are produced in the gland cells and
secreted into the blood or lymph to exercise control over metabolism and development of the orga-
nism.
The following general biological signs are characteristic of hormones:
- Remote action, i.e. the hormones control metabolism and the functions of effector cells at a
distance.
- Strict specificity of biological action, i.e. functionally, no hormone can be entirely replaced by
another one.
- High biological activity, i.e. quite small amounts of hormone, in some instances of the order of
10
-5
g, are sufficient to provide for the vital activity of the organism.
96
The hormone-secreting glands are occasionally subdivided into the central glands (anatomically
related to specialized portions of the central nervous system) and the peripheral glands.
Endocrine functions are also exercised by other cells capable of secreting biologically active com-
pounds whose properties resemble those of hormones. For this reason, these agents are conventionally
referred to as hormone-like compounds, or hormonoids (local hormones, parahormones). Their action
is, a rule, confined to the site where they are produced; they are secreted by cells dispersed about diffe-
rent tissues. Hormonoids are secerted by the cells of gastrointestinal tract (they control digestion), by
the intestinal enterochromaffin cells (which produce serotonin a regulator of intestinal function), by
the mast cells of connective tissue (heparin and histamine), and by the cells of kidneys, seminal vesi-
cles, and other organs (prostaglandins). Presumably, these endocrine cells are specific peripheral rep-
resentatives of the endocrine system. This is borne out by the occurrence of certain true hormones in
the peripheral organs, for example, somatostatin in the pancreas and liver.
By their chemical structure, hormones are subdivided into:
1. Protein-peptide hormones (those of hypothalamus, pancreas, pituitary and parathyroid glands,
thyroidal calcitonin).
2. Amino acid derivatives (adrenalin, derived from amino acids phenylalanine and tyrosine; iodo-
thyronines, derived from tyrosine; melatonin, derived from tryptophan).
3. Steroids (sex hormones: androgens, estrogens, and gestagens; corticosteroids).

Neuroendocrine relationship scheme
Information on the state of internal and extaernal media of the organism is transmitted to the ner-
vous system to be processed therein; in response, regulatory signals are transmitted to the peripheral
tissues and organs. The production and secretion of hormones by the peripheral glands are accompli-
shed incessantly. This is essential for maintaining the hormones at appropriate level in the blood, since
they are liable to fast inactivation and elimination from the organism. The required level of a hormone
in the blood is maintained owing to the self-regulation mechanism. This mechanism is based on inter-
hormonal relationships which are referred to as plus-minus interactions, or mutually exclusive rela-
tionships.
Hormonal control: general outline
Hormones, which are secreted by the glands into the blood, usually become bound to specific
blood plasma transport proteins (or, in certain cases, adsorbed on blood cells) to be carried to the peri-
pheral tissues, where they exert influence on metabolism and tissue function. Various tissues respond
to hormonal action differently. Tissues (cells, organs) exhibiting a high sensitivity to hormones, i.e.
the tissues in which the hormones elicit pronounced charges in metabolism and function, are called
targets for a given hormone, while other, less responsive, tissues are assigned to nontarget tissues;
it should be admitted that this classification is rather arbitrary. The hormonal influence extends, in a
wider sense, to all metabolic manifestations. However, in order to get a better understanding of the
mechanism of action of any hormone or, in fact any extracellular regulator, it is worthwhile to consi-
der the general principles of hormonal metabolic control.
The following possible types of action for extracellular regulators (often referred to as first mes-
sengers), including hormones, are distinguished:
- membrane, or local, action,
- membrane intracellular, or indirect, action,
- cytosolic, or direct, action.
Membrane type of hormonal action. The membrane type of hormonal action consists in that the
hormone, at the site of its binding with the cell membrane, renders the membrane permeable to gluco-
se, amino acids, and certain ions (Ca
2+
, K
+
). In this case, the hormone acts as an allosteric effector for
membrane transport systems. The glucose and amino acids supplied exert, in turn, influence on bio-
chemical cellular processes, while a change in ion partition on both sides of the membrane affects the
electric potential and function of the cell. As a typical example, insulin may be cited which exhibits
both a membrane type (by eliciting local changes in the transport of certain ions, glucose, and, possi-
bly, amino acids) and a membrane-intracellular type of action.
Membrane-intracellular mechanism for metabolic control. The membrane-intracellular type of
action in characteristic of hormones or functionally related to them extracellular regulators called first
messengers. These are not capable of entering the cell and cannot therefore influence the intracellular
processes directly. Their action is effected indirectly, through the agency of an intracellular mediator
97
(or second messenger) which triggers a chain of successive biochemical reactions leading to a modifi-
cation of cellular functions.
The sequence of information transfer from diverse external stimuli into the cell may be outlined
within the following general scheme: external signal (first messenger) membrane receptor trans-
ducer chemical amplifiersecond messengerinternal effectorcell response.
The first messenger (for example, a hormone) becomes bound to the receptor on the outer side of
plasma membrane. The membrane receptors are a kind of start buttons on a control desk of cellular
metabolism: the hormone receptor complex thus formed acts on a protein, the membrane transducer,
which, in turn, transmits the signal to an enzyme (chemical amplifier) acting as a catalyst for the
production of a second messenger inside the cell. The second messenger binds to a special protein
(internal effector) which exerts influence on the activity of definite enzymes or on the properties of
nonenzymic proteins conducive to alterations of cellular processes, viz. chemical conversion rates,
permeability, secretion, structural contractility, activity of genes, etc.
There are known several groups of compounds that are candidates for the function of a second
messenger. These are:
1. Nucleotides: cyclic nucleotides (3,5-cAMP and 3,5-cGMP), 2,5-oligo(A)n (adenyl oligo-
nucleotide).
2. Lipids: diacylglyceride, inositol triphosphate.
3. Ca
2+
ions.
4. Peptides.
Metabolic control effected via cyclic nucleotides. The intracellular regulators affect the produc-
tion of cyclic nucleotides via either of the two signal systems: adenylate cyclase or guanilate cyclase.
The former controls the production of cAMP and the latter of cGMP.
Adenylate cyclase is built into the membrane. This enzyme catalyzes the production of cAMP ac-
cording to the scheme:
ATP 3,5-cAMP + H
4
P
2
O
7

The interaction of the external regulator with R-type receptors brings about a conformational
change in the latter, and this effect is transmitted to the G-protein which thus acquires an ability to
bind GTP. The G-GTP complex acts as an allosteric activator towards adenylate cyclase. Its activation
initiates the production of cAMP from the intracellular ATP. Thus, at the moment as the G-GTP com-
plex is formed, information is transferred from the external signal molecule to an internal molecule,
the second messenger. Until the G-GTP complex persists, adenylate cyclase continuously catalyzes the
cAMP synthesis, owing to which the external chemical signal becomes multiply amplified: per one
molecule of R-bound first messenger, as many as 100-1000 molecules of cAMP, the second messen-
ger, are generated.


The production of cAMP ceases as the G-GTP complex becomes inactivated, which is effected by
the action of GTPase acting to hydrolyze GTP to GDP in this case, G loses its ability to activate ade-
nylate cyclase. The cholera toxin is known to inhibit GTPase extending thereby the lifespan of G-GTP
complex which elicits the continuous production of cAMP. In the intestinal cells, cAMP makes the
98
secretion of fluid in the organism sharply increase, which explains the acute diarrhea in patients with
cholera.
The othere cyclic nucleotide, a potential candidate for the second messenger, is cGMP. It is pro-
duced from GTP with the participation of guanylate cyclase. In distinction from adenylate cyclase, this
enzyme is loosely bound to the plasma membrane.
Further transmission of the signal through the aid of cAMP and cGMP is effected with the partici-
pation of protein kinases as internal effectors. The protein kinase function is phosphorylation of defini-
te proteins according to the scheme: protein +ATP phosphoprotein +ADP.
Since the cyclic nucleotides transmit information to protein kinase which is catalyst, an added,
10-to-100-fold increase in chemical signal occurs, otherwise stated, each of the protein kinase molecu-
les, while persisting in an activated state, is able to phosphorylate and, therefore, to functionally modi-
fy, to about a hundred diverse proteins. Thus a single hormone molecule is potentially capable of mo-
difying the structure and affecting the function of nearly a million molecules in the cell.
In the cell are systems that offset the influence of cyclic nucleotides on biochemical processes.
The hydrolysis of cyclic nucleotides is carried out by phosphodiesterases according to the scheme:

3,5-cAMP phosphodiesterase + H
2
O 5-AMP
or 3,5-cGMP phosphodiesterase + H
2
O 5-GMP

The 5-AMP and 5-GMP thus formed are incapable of activating the protein kinases. Numerous
regulators act to increase cAMP concentration. They encompass all the hypothalamic and hypophyseal
hormones, calcitonin, parathyrine, glucagon, catecholamines (acting via |-adrenergic receptors), vaso-
pressin, histamine, and certain prostaglandins. The action exerted by hormones is reversible; it is ope-
rable within a few seconds or minutes after the contact of hormone with a membrane receptor. The
hormones whose action is mediated by cAMP produce not only short-term metabolic changes (i.e. sti-
mulate lipolysis, glycogenolysis, ion transport, etc.), but long-term ones also. Caffeine and theobromi-
ne are the competitive inhibitors of enzyme phosphodiesterase.


Caffeine Theobromine

Metabolic control via Ca
2+
ions. The intracellular concentration of Ca
2+
ions is negligeably small
10
-7
mol/L, while outside the cell, their concentration is as high as 10
-3
mol/L. The Ca
2+
ions are deli-
vered into the cell from the external medium via several calcium channels in the cell membrane.
The Ca
2+
ions flow is controlled by the Ca
2+
-ATPase of cell membrane which pumps Ca
2+
ions out at
the expense of ATP energy. The elimination of Ca
2+
ions from the intracellular depots is accomplished
in a manner similar to their supply from the exterior via calcium channels. The calcium supplied from
the external medium or from intracellular depots by the action of various external stimuli interacts
with cytoplasmic calcium-binding proteins. As has been mentioned earlier, the most important of them
is cadmodulin (CaM).
The Ca
2+
-CaM complex by activating the protein kinase sensitive to it or other enzymes exerts
control over biochemical processes and other functional activities of the cell related to them.
Cell-invading drugs inhibit either of the phosphodiesterases cAMP or cGMP, they may simulate a
metabolic effect due to natural hormones and mediators operative via cyclic nucleotides. The phospho-
diesterase activators inhibit the action of these hormones and mediators. This possibility is made use
of in practical medicine by applying drugs that can inhibit phosphodiesterase. These include xantine
derivatives, such as caffeine, theophylline, theobromine, and euphylline. These drugs mainly increase
the concentration of cAMP, while such a drug as trental increases chiefly the cGMP concentration.
Cytosolic mechanism of hormone action. Cytosolic mechanism is typical of the hormones that
can penetrate through the lipid layer of plasmatic membrane, i.e. are related by their physico-chemical
properties, to lipophilic compounds, for example, steroid hormones (vitamin D, by its metabolic effect,
is also close to steroid hormones, and for this reason it is often assigned to hormonal agents).
99
Hormones with cytosolic type of action penetrate the cell to be complexed with cytosolic or nuclear
receptors. The receptor-complexed hormones control the enzyme concentration in the cell by
selectively affecting the gene activity of nuclear chromosomes and exerting thereby influence on
metabolism and functions of the cell. Since the cell-invading hormone is involved in the mechanism of
enzyme concentration control, this type of action is called direct action in contrast to the membrane
intracellular mechanism, when a hormone controls metabolism indirectly, through the agency of
second messengers. In terms of lipophilicity, iodothyronines are found midway between steroids and
other water-soluble hormones. Probably, for this reason they exhibit a combined type of action on cell
metabolism, i.e. both membranes intracellular and cytosolic.
Production and practical application of hormones. For their practical applications, hormones are
produced by extraction from biological materials, by chemical synthesis, or by genetic engineering
methods. The first of the above techniques is employed in the production of insulin and glucagon from
bovine pancreas, of corticotropin and melanotropin from bovine pituitary, of folitropin (serum gonado-
tropin) from blood serum, and of lutropin (chorionic gonadotropin) from the urine of pregnant mares.
Currently, chemical synthesis is widely used for production of all steroid hormones, their analogues
and derivatives iodothyronines and other hormones-amino acid derivatives and peptide hormones of
oxytocin type. Practically all the protein hormones can be prepared by laboratory synthesis. Insulin,
somatostatin and related hormones have been prepared by the genetic engineering method in the labo-
ratory.
In medical practice, hormones are used for substitution therapy and pathogenetic therapy. In the
latter instance, certain specific properties of hormones (antiinflammatory, anabolic, etc.) are made use
of, even through the hormonal concentrations in the patients organism may be at normal level.

LECTURE 24

Subject: HORMONES OF HYPOTHALAMUS-HYPOPHYSEAL SYSTEM.
HORMONES OF PERIPHERAL GLANDS

Production and secretion of hormones by hypothalamus and hypophysis are closely related and
for this reason it appears expedient to discuss the relevant processes in parallel. In the anterior lobe of
the pituitary gland (adenohypophysis), tropic hormones are produced, while the posterior pituitary lobe
(neurohypophysis) releases only neurohormones (vasopressin and oxytocin), which are produced in
the hypothalamus nuclei.
By their chemical structure, thyrotropin, follitropin, and lutropin are glycoproteins. They are com-
posed of two subunits, o and |. In all these glycoproteins, the o-subunits are identical, while the |-su-
bunits are structurally different and determinative of the specificity of hormonal action. Other relevant
hormones are simple proteins possessing a single polypeptide chain; vasopressin and oxytocin are cyc-
lic octapeptides.
The secretion of tropic hormones is controlled by hypothalamic peptides. To date the following
hypothalamic neuropeptides, which are regulators of hypophyseal hormonal secretion, have been iso-
lated:
Tropic (pituitary) hormone Hypothalamic hormone
Somatotropin Somatoliberin, somatostatin
Corticotropin Corticoliberin, corticostatin
Thyrotropin Thyroliberin, thyrostatin
Follitropin Folliberin
Lutropin Luliberin
Prolactin Prolactoliberin, prolactostatin
Melanotropin Melanoliberin, melanostatin

Mechanism of action and functions of hypophyseal hormones
All the tropic hormones exert their action either on the function of peripheral glands, or directly
on the peripheral tissues by binding to the membrane receptors and by activating adenylate cyclase.
cAMP effects the hormone production or metabolism in the target cells.
The effects that the hypophyseal hormones can produce may be divided into four groups:
100
1. Control of biosynthesis and hormonal secretion by peripheral glands (thyrotropin, follitropin,
lutropin, corticotropin, and somatotropin).
2. Control of sex cell production (follitropin).
3. Functional and metabolic control of effector tissues and organs (somatotropin, o- and |-lipo-
tropins, corticotropin, lutropin, follitropin, melanotropin, prolactin, oxytocin, and vasopressin).
4. Functional control of the nervous system (corticotropin, |-lipotropin, and others).

Direct effect of hypophyseal hormones on peripheral tissues
Corticotropin exerts a direct action on fat tissue by stimulating the tissues glucose absorption
and release of fatty acids and glycerol. The hormonal fat-mobilizing effect is related to adenylate cyc-
lase activation; the cAMP formed stimulates triacylglyceride lipase, which splits triacylglycerides into
glycerol and fatty acids. In addition, the hormonal action of corticotropin on melanin production and
skin pigmentation is similar to that of melanotropin.
o- and |-lipotropins exert a specific fat-mobilizing action by the mechanism typical of all c-
AMP-stimulating hormones.
Gonadotropins produce a fat-mobilizing effect similar to that of lipotropins. Moreover, prolactin
stimulates protein biosynthesis and lactose production by the mammary gland epithelium.
Vasopressin, or antidiuretic hormone, in addition to its fat-mobilizing action, exerts a selective
control of water reabsorption in the distal tubes and collecting ducts of the kidneys and activates ade-
nylate cyclase. cAMP activates protein kinases, which phosphorylate the cell membrane proteins to in-
crease their permeability for water. The water reabsorption reduces diuresis, makes increase of the uri-
ne density and urinary concentrations of sodium and chlorides. Vasopressin stimulates the contraction
of the muscular tissue of the capillaries and arterioles and produces a moderate rise in blood pressure.
Deficiency in vasopressin develops a disease called diabetes insipidus. It is manifested by a large
discharge of urine (4 to 10 liters per day) of low density (1.002-1.006); excessive thirst (polydipsia)
develops. Administration of vasopressin preparations removes the symptoms of this disease.
Oxytocin stimulates contraction of uterine muscles; this action is associated with an increased
intracellular Ca
2+
concentration and with cGMP production. Oxytocin accelerates protein synthesis in
the mammary glands during the lactation period (in part, this effect is also exhibited by vasopressin)
and stimulates the release of breast milk owing to an enhanced contractile activity of the milk duct
myoepithelium. This hormone produces an insulin-like effect on the fat tissue, i.e. increases glucose
consumption and triglyceride synthesis in it.
Melanotropin. o- and |-melanotropins are distinguished. The two hormones are secreted by the
anterior pituitary and affect the production of melanin in skin, iris of the eye, and the epithelial pig-
ment of the retina. They produce a fat-mobilizing action on the fat tissue by stimulating cAMP genera-
tion in it.
Somatotropin, or growth hormone, is the only hormone that exhibits a biological species-specific
effect. The animal somatotropin produces no effect on the humans. It also controls the growth of inter-
nal organs and soft tissues of the face and oral cavity. Its direct action is associated with the activation
of adenylate cyclase and formation of cAMP, for example, in muscles and insular pancreatic tissue.
Somatotropin deficiency in the young organism is conducive to a premature growth cessation,
with on eventual development of dwarfism. The stature of an adult dwarf is 100-120 cm. Hypophyseal
dwarfism in distinction to those afflicted with hypothyroid dwarfism, has a proportionate constitution,
with no signs of mental retardation. Somatotropin hypersecretion in a juvenile period shows up as gi-
gantism; in a maturity period, it is conductive to a state called acromegalia. Acromegalia is manifested
by an enlargement of the prominent parts of the face (nose, chin, superciliary eyebrows).

Hormones of peripheral endocrine glands
Thyroidal hormones. The thyroid gland secretes hormones of two groups, which affect metabo-
lism differently. I odothyronines-thyroxineand iodothyronine-control energy metabolism and exert in-
fluence on cell division and differentiation, determining thereby the development of the organism.
Calcitonin (polypeptide hormone with a molecular mass of about 30,000) controls phosphorus-cal-
cium metabolism: it appears more expedient to discuss calcitonin action in parallel to that of parathy-
roid glands.
Iodothyronines exert action on numerous tissues of the organism, the highest sensitivity being
exhibited by the tissues of liver, heart, kidney, skeletal muscles, and, to a lesser extent, adipose and
nervous tissues. In the organism, the thyroid hormones affect to a higher degree cell division, cell dif-
101
ferentiation, and energy metabolism. Alterations in energy metabolism due to the so-called calorigenic
property of thyroid hormones; outwardly show up as an increased oxygen consumption and production
of heat. Iodothyronines exert influence on metabolism via cytosolic receptors by effecting the nuclear
chromosomes, and via cAMP.
For the synthesis of iodothyronines essential are iodide as supplied by active transport from blood
to thyroidal epithelium and thyroglobulin which is produced in the epithelium and fills the follicular
cavity to form colloid. Iodothyronine synthesis proceeds by several steps which are:
1. Formation of an active iodine from iodide through the aid of iodide peroxidase by reaction:
I
-
- e (2e) I
-

H
2
O
2
serves as an electron acceptor. The active iodine is capable of iodinating tyrosine.
2. Iodination of tyrosine constitutive of thyroglobulin with the participation of tyrosine iodinase.
The products formed are monoiodotyrosine or diiodotyrosine.
3. Oxidative condensation of mono- and diiodotyrosines with the formation of triiodothyronine
and tetraiodothyronine (thyroxine) within the thyroglobulin molecule. The process is carred out on the
surface of tyrosine iodinase.
4. Uptake of thyroglobulin from the colloid by epithelium cells and its translocation to the outer
membrane surface bathed by the extracellular fluid. This process is reminiscent of endocytosis.
5. Secretion of iodothyronines, which is accomplished by hydrolysis of thyroglobulin by protea-
ses enabling the release of thyroxine (T
4
) and triiodothyronine (T
3
) into blood.


Figure 57. Chemical structure of thyroxine (T
4
)

The thyroid gland malfunction may entail an excess or deficiency of iodothyronines in the orga-
nism. In the hyperfunction of thyroid gland, or hyperthyroidism, excessive production of iodothyroni-
nes is observed. Acute hyperfunctional forms have been named thyrotoxicosis, or Basedows disease,
since the symptoms of disturbed metabolism and malfunction may be likened to iodothyronine intoxi-
cation. In hypofunction of the thyroid gland, or hypothyrosis, the organism is in short supply of iodo-
thyronines. Hypothyrosis in neonates or infants is called cretinism, or infantile myxedema; in adults, it
is called simply myxedema.
Hormones of parathyroid glands. The parathyroid glands secrete two protein hormones calcito-
nin (also secreted by the thyroid gland) and parathyrin (parathormone). Calcitonin and parathyrin
control the balance of Ca
2+
ions and inorganic phosphate in the organism. In turn, the secretion of cal-
citonin and parathyrin, which lack appropriate tropic hormones, is feedback-controlled by Ca
2+
ions.
An increased concentration of Ca
2+
ions in blood plasma elicits the secretion of calcitonin, on the con-
trary, a decreased percentage of Ca
2+
in blood plasma is a stimulus for liberation of parathyrin from the
glands. Parathyrin to a significant extent exerts influence on phosphorus-calcium metabolism through
vitamin D.
Hormones of pancreatic gland
Hystologically, in the pancreas distinguished are insular (Sobolev-Langerhans islands) and acino-
us tissues which contain cells responsible for synthesis and secretion of a number of hormones. The
cells of A-type (o-cells), B-type (|-cells), and D-type (all of them contained in insular tissue) secrete,
respectively, glucagon, insulin and somatostatin.
Glucagon is a protein with M.m. of 3,485; it is composed of 29 amino acids. It binds to the mem-
brane receptors of target tissues. The targets for glucagon are liver, fat tissue, and, to a lesser extent,
muscles. By activating adenylate cyclase and by making increase the cAMP concentration, glucagon
elicits mobilization of glycogen in liver and, partly, in skeletal muscles, and triacylglycerides in fat tis-
sue. Mobilization of these energy reserves leads to increased levels of glucose, fatty acids, and glyce-
rol in blood.
I nsulin is formed in the |-cells as preproinsulin which, when subjested to hydrolysis, yields pro-
insulin. Proinsulin, containing 84 amino acid residues, suffers the cleavage of a polypeptide fragment
(called C-peptide) composed of 33 amino acids. This results in the formation of insulin (of a molecular
mass of about 6,000) composed of 51 amino acid residues. Insulin has two chains: a short one (A-
102
chain) made up of 21 amino acids and a long one (B-chain), of 30 amino acids. The two chains are
cross-linked through disulphide bridges.
It supplied to the blood occurs in a free state or as bound to plasma proteins. Free insulin exerts
influence on the metabolism of all insulin-sensitive tissues, while the bound insulin, on fat tissue only.
The insulin-sensitive tissues include muscular and connective tissues. Membrane insulin receptors of
glycoprotein nature have been found in the tissues. The insulin-receptor complex is capable of drasti-
cally changing the cell membrane permeability for glucose, amino acids, Ca
2+
, K
+
, and Na
+
ions, or to
be more precise, of accelerating the transport of glucose, amino acids, and Ca
2+
and K
+
ions into the
cell.
The excessive insulin may be observed in tumoral islands (insulomes) or in overdosed insulin the-
rapy. With deficient insulin, a widely spread disease, diabetes mellitus, develops. Diabetes mellitus de-
velops under true and false insulin deficiency. The true insulin deficiency is due to a disorder in pro-
duction and secretion of insulin in the insular |-cells (insulin-depending diabetes mellitus, type I). In
patients with deficiency of receptors to insulin disease called insulin-independent diabetes mellitus, ty-
pe II.
In diabetes mellitus take place the changes in carbohydrate, lipid, protein, and water-mineral me-
tabolisms. The metabolic disturbances are: hyperglycemia and glucosuria, hyperaminoacidemia and
hyperaminoaciduria, increased concentrations of fatty acids, glycerol, and cholesterol in blood, and ke-
tonemia, and ketonuria.
Insulin preparations are used in the therapy of diabetes mellitus, as well as anabolic stimulators in
dystrophy of organs.

Hormones of the adrenal glands
Hormones of adrenal medulla. In the medulla of the human adrenal glands, adrenalin and, to a
lesser extent, noradrenalin, are formed.


Figure 58. Chemical structure of noradrenalin (left) and adrenalin

Adrenalin exerts a dual effect on the metabolism of target tissues depending on the predominant
occurrence in them of either o-, or |-adrenoreceptors to which the hormone becomes bound. The bin-
ding of adrenalin to |-adrenoreceptors stimulates adenylate cyclase and produces metabolic alterations
characteristic of cAMP. The adrenalin binding with o-adrenoreceptors stimulates guanylate cyclase
and produces metabolic alterations typical of cGMP. The alterations that occur in carbohydrate and
lipid metabolisms are very much the same as those produced by glucagon.
Adrenal cortex hormones. In the adrenal cortex, steroid hormones, or corticosteroids, are formed
from cholesterol. Corticosteroids, by the physiological effect they produce, are subdivided into three
groups:
- glucocorticoids chiefly affecting carbohydrate metabolism,
- mineralocorticoids chiefly affecting mineral metabolism,
- sex hormones (male hormones androgens, and female hormones estrogens).
The human adrenal glands secrete glucocorticoids (hydrocortisone and corticosterone) and a mi-
neralocorticoid (aldosterone).

Figure 59. Chemical structure of corticosteroids: cortisol (left) and aldosterone

103
The secretion of glucocorticoids is controlled by corticotropin. Glucocorticoids (cortisol) become
bound to o1-globulin of blood plasma, called transcortin, to be transported in the complexed stat to
peripheral tissues. The targets for glucocorticoids are liver, kidney, lymphoid tissue (spleen, lymph no-
des, lymphoid plaques of the intestine, lymphocytes, and thymus), connective tissues (bones, subcuta-
neous connective tissue, and adipose tissue), and skeletal muscles. These tissues contain cytosolic re-
ceptors for binding glucocorticoids. To be noted, a hormone-cytoreceptor complex may exert an enti-
rely opposite effect on protein synthesis in different tissues. In the liver and kidneys, glucocorticoids
favour the utilization of amino acids in gluconeogenesis, since they act as specific inducers of the syn-
thesis of gluconeogenesis enzymes.
Since glucocorticoids enhance the secretion of adrenalin from the adrenal medulla, the action of
glucocorticoids becomes augmented by the metabolic effect of adrenalin. Thus, glucocorticoids mo-
bilize triacylglycerides from the adipose tissue at the expense of adenylate cyclase activation, although
the membrane intracellular activity is not typical of them. Apparently, the mobilization of fat from the
fat depots is associated with adrenalin. As a result, glycerol and fatty acids are supplied to the blood;
glycerol is used in gluconeogenesis, while fatty acids are consumed in the liver to produce ketone
bodies which are excreted in the blood.
Glucosuria, aminoaciduria, and ketonuria set in, as the concentrations of glucose, amino acids,
fatty acids, glycerol, and ketone bodies in the blood increase. On the whole, these metabolic changes
resemble a picture of diabetes mellitus. This diabetic state is of different nature and for this reason is
referred to as steroid diabetes.
Glucocorticoids produce changes in the water-salt metabolism: they increase the Na+ ion reab-
sorption and renal excretion of K+. They return sodium and water in the extracellular space of the
organism tissues (which may lead to oedemas). This action is similar to the effect due to mineralocor-
ticoids, only less pronounced. Inhibition of the bone tissue protein synthesis leads to local deossifica-
tion of the bones. Calcium and phosphorus are eliminated from the affected bone tissue into the blood
and then are excreted in the urine.
Glucocorticoids and their analogues are widely applied in clinics. The medicinal effect of gluco-
corticoids is based on their ability to affect lymphoid and connective tissues. Glucocorticoids inhibit
the formation of antibodies in the lymphoid tissue to produce the state of sensitization towards inva-
ders, and thus prevent further development of allergic response and inflammation.
Mineralocorticoids (aldosterone) controls the balance of Na
+
and K
+
cations. At low Na
+
and
high K
+
concentrations in blood, the synthesis and secretion of aldosterone become increased.
Aldosterone controls the balance of Na
+
, K
+
, Cl
-
ions and water in the organism; for this reason,
the normal function of this hormone is of utmost importance for the vital activity of the organism.
Aldosterone is transported in the blood to tissues by using plasma albumins as carrier adsorbents.
The targets for aldosterone are epithelial cells of distal tubules of the kidney, which contain a large
number of cytoreceptors for binding this hormone. The aldosterone-cytoreceptor complex penetrates
the nuclei of the renal tubule cells and activates the transcription of chromosomal genes that carry in-
formation on the proteins involved in the transport of Na
+
ions across the membranes of tubular epi-
thelium. Owing to this, the reabsorption of Na
+
and its counterion Cl
-
from the urine into intercellular
fluid and further into blood becomes increased. Simultaneously, K
+
ions are excreted (in exchange for
Na
+
) in the urine from the epithelium of renal tubules. On the whole, the aldosterone effect is manifes-
ted by the retention of Na
+
, Cl
-
, and water in the tissues and by urinary loss of K
+
ions.
Disturbances of hormonal function of the adrenal glands. In such forms of hypercorticoidism,
or Cushings disease (which occurs due to the impaired hypothalamohypophyseal system conducive to
corticotrophin hypersecretion) and corticosteroma (a tumor active chiefly in the synthesis of hydrocor-
tisone), a hyperproduction of glucocorticoids is observed.
There occurs hypercorticoidism attended by excessive secretion of aldosterone (hyperaldostero-
nism, or Konns disease). In this disease, the symptoms of influence of aldosterone excess on the wa-
ter-salt balance are observed, viz. oedemas, high blood pressure, and myocardial hyperexcitability.
Hypocorticoidism, also called Addisons disease, or bronze disease, is manifested by a deficiency
in all the corticoids and attended by manifold alterations in metabolism and functions of the organism.

Sex hormones
The sex glands (gonads) are paired organs represented by the testes in males and by the ovaries in
females. The male sex hormones, androgens, are produced by Leidigs cells, and the female sex hor-
104
mones (estrogens) are produced in the ovarian follicles. The male and female hormones are synthesi-
zed from their common precursor, cholesterol.
Female sex hormones, estrogens (estradiol) and progesterone, have the cytosolic mechanism of
action. Most physiological effects due to estrogens are basically determined by the hormonal influence
of estrogens on the activity of specific chromosomal genes. These effects ultimately induce the synthe-
sis of specific proteins which are decisive for characteristic alterations in metabolism, cell growth, and
cell differentiation. The pronounced anabolic action of estrogens, i.e. their ability to stimulate protein
synthesis in target organs, provides for a positive nitrogen balance in the organism. Estrogens are
inducers of glycolysis and pentose phosphate cycle. The estrogen hormones accelerate the renewal of
lipids, inhibit accumulation of lipids in liver and fat tissues, favour the elimination of cholesterol from
the organism and lower the cholesterol level in blood.
Progesterone exerts physiological and biochemical effects only during the luteal phase of ovarian
cycle.


Figure 60. Chemical structure of sex hormones: estradiol (left) and testosterone

Androgens (testosterone), prior to be supplied to the tissues become bound to glycoprotein of
blood plasma called testosterone-estradiol-binding globulin (which is a specific binder for testosterone
and estradiol). The androgens become bound to androgenic receptors and act on the nuclear chromatin
of target cells to facilitate DNA synthesis activation during replication and to accelerate the specific
gene transcription. The anabolic action of testosterone is much superior to that of estrogens. In males,
this results in the development of powerful skeletal musculature in the development and mineraliza-
tion of the epiphyseal growth zones of the bones.
Testosterone preparations and their synthetic analogues, anabolic steroids, are used clinically.
Anabolic steroids (methylandrostenediol, nerobolil, retabolil) are used to treat various dystrophies,
diabetes mellitus. Female sex hormones are also used clinically.
Antihormones are compounds that exhibit antihormonal activity through binding to cytosolic re-
ceptors. While being active antagonists towards other hormones, they may either exhibit or not the
intrinsic hormonal activity. The molecular mechanism of antihormonal action is based on the competi-
tion for binding sites with the corresponding cytosolic receptors. Owing to this, the complex antihor-
mone-cytosolic receptoris incapable of acting as a protein synthesis inducer in the cells. An antihor-
mone displaces a true hormone only when its concentration in the cell is very high.
Natural antihormones include estrogens and androgens which complete with each other for bin-
ding with the receptors of opponent hormone: estrogens block the androgenic receptors, and andro-
gens, the estrogenic ones.
Antihormones are used in treating hormone-dependent tumors when a need arises to prevent the
action of hormone on proliferating tumoral cells, in therapy of abnormal sexual behaviour (hyperse-
xuality), etc.
Prostaglandins are hormone-like compounds (hormonoids) derived from C
20
-polyene fatty acids
containing a cyclopentane ring (see Lecture 15 Biosynthesis of lipids). Prostaglandins are short-
lived species that are synthetized at heed in small amounts to exert a local biological effect at the site
of their formation. An excessive production of prostaglandins or their deficiency may lead to patholo-
gic processes such as inflammation, thrombosis, gastric ulcer, and others.

LECTURE 25

Subject: BIOCHEMISTRY OF BLOOD: CHEMICAL COMPOSITION OF
BLOOD PLASMA
Blood is fluid tissue composed of cells (formed elements of the blood) and an extracellular liquid
105
medium. The overlying liquid (supernatant) of blood sample obtained on precipitation of the blood
cells in the presence of an anticoagulant is called blood plasma. The plasma is an opalescent liquid
containing all extracellular components of the blood. The blood cells account for about 45%, and the
plasma, for about 55% of the blood volume. The clear liquid that separates from the blood when it is
allowed to clot completely is called blood serum. Actually, the blood serum is the plasma from which
fibrinogen has been removed in the process of clotting.
Water accounts for about 83% of blood accounts for the rest. By its physico-chemical properties,
blood is a viscous liquid of specific density 1.050-1.060. The blood viscosity and density are depen-
dent on the relative contents of blood cells and plasma proteins. The blood pH value (7.36-7.44) is
maintained by buffer systems at a constant level, its variations not exceeding 0.05-0.10 pH unit.
Blood components. Cellular and extracellular blood components are different origin, and for this
reason they may be regarded as indicators of biochemical physiological processes occurring in the
tissues and organs from which they are supplied to the blood. Alterations in the blood composition are
therefore diagnostic of the state of health of the human organism.
Alterations in the quantitative and qualitative composition and in biochemistry of the blood cells
are warning signals on disturbances in the bone marrow; any abnormality in lymphocytes is indicative
of an impaired function of both bone marrow and lymphoid tissue. Most chemical components of blo-
od plasma are supplied from various organs. Since the cell membranes are impermeable or poorly per-
meable to macromolecules (proteins and enzymes), macromolecules are secreted from tissues and
organs actively (by exocytosis). Major sources of macromolecules are liver, endocrine glands, partial-
ly intestinal mucosa, and blood cells themselves. Under physiological conditions, other organs and tis-
sues are but to a small extent involved in the generation of blood plasma proteins. Low-molecular
plasmic components are of more diversified origin and specify various aspects of tissue metabolism.
Biochemical and physiological functions of the blood are defined by a cooperative participation
of blood cells and chemical components of blood plasma in metabolism.
Biochemical functions of blood and their characterization. The blood performs the following
functions:
- transport,
- osmoregulatory,
- buffering,
- detoxifying,
- defensive, or immunologic,
- regulatory, or hormonal,
- hemostatic.
Chemical composition of blood plasma
I . Proteins
1. Total protein 65-85 g/L
2. Albumins 35-50 g/L
3. Globulins 25-35 g/L
4. Fibrinogen 2.0-7.0 g/L
5. Haptoglobin 0.28-1.90 g/L
6. Prothrombin 10-15 mg/dL
7. Plasminogen 1.4-2.8 mol/L (20-40 mg/dL)
8. Transferrin 19.3-45.4 mol/L (170-400 mg/dL)
9. Ceruloplasmin 1.52-3.31 mol/L (23-50 mg/dL)
10. |-Lipoproteins 3.0-6.0 g/L (300-600 mg/dL)
HDL high density lipoprotein (o-LP) 1.063-1.210 mmol/L (80-400 mg/dL)
LDL low density lipoprotein (|-LP) 1.006-1.063 mmol/L (360-640 mg/dL)
I I . Enzymes
1. Alanyl aminotransferase (ALT) 0.16-0.68 mmol/h L or (15-75 IU/L)
(glutamate pyruvate transferase, GPT)
2. Aspartate aminotransferase (AST) 0.10-0.45 mmol/h L or (10-50 IU/L)
(glutamate oxaloacetate transferase, GOT)
3. Lactate dehydrogenase 0.8-4.0 mmol/h L
4. Creatine kinase < 1.2 mmol/h L or (< 90 IU/L)
5. Fructose-biphosphate aldolase (F-1,6-PA) 3.6-21.8 mmol/h L
106
6. Acetylcholine esterase 160-340 mmol/ h L
7. o-Amylase 15-30 g/h L or (< 300 IU/L)
8. Alkaline phosphatase 30-150 IU/L
9. Acidic phosphatase < 62 nkat/L
10. -Glutamyl transferase (GT or GGT) < 60 IU/L
I I I . Nonproteinic nitrogenous compounds
1. Nitrogen residual (nonproteinic) 19.5-30.0 mmol/L
2. Nitrogen of amino acids 3.5-5.5 mmol/L
3. Creatine 15-70 mmol/L
4. Creatinine 60-150 mol/L
5. Urea 3.3-6.7 mmol/L
6. Uric acid 0.1-0.4 mmol/L
7. Bilirubin total 8-20 mol/L
8. N-Acetylneiraminic acid 1.8-2.2 mmol/L
9. Histamine 17.99-71.94 nmol/L (0.2-0.8 g/dL)
10. Adrenalin 1.91-2.46 nmol/L (0.35-0.45 g/L)
11. Serotonine 0.3-1.7 mol/L (5.0-30.0 g/dL)
12. Thyroxine 64.36-141.59 nmol/L (5-11g/dL)
I V. Carbohydrates and metabolites
1. Glucose 2.8-6.0 mmol/L
2. Lactate 0.5-2.0 mmol/L
3. Pyruvate < 0.1 mmol/l
4. Citric acid 88.5-156.1 mol/L (1.7-3.0 mg/dL)
V. Lipids and metabolites
1. Total lipids 4.0-8.0 g/L
2. Triacylglycerides 0.5-2.1 mmol/L
3. Total phospholipids 2.0-3.5 mmol/L
4. Total cholesterol 4.0-8.6 mmol/L
5. Free fatty acids 0.3-0.8 mmol/L
6. Ketone bodies 100-600 mol/L
VI . Mineral components
1. Sodium (Na
+
) 135-155 mmol/L
2. Potassium (K
+
) 3.6-5.0 mmol/L
3. Chlorides (Cl
-
) 97-108 mmol/L
4. Calcium (Ca
2+
) 2.25-2.75 mmol/L
5. Phosphate inorganic 0.8-1.4 mmol/L
6. Magnezium (Mg
2+
) 0.7-1.0 mmol/l
7. Sulphates 0.4-0.6 mmol/L
8. Iron (Fe) 14-32 mol/L (65-175 g/dL)
9. Copper (Cu) 12-19 mol/L
10. Zinc (Zn) 12-20 mol/L
11. Ammonia 10-47 mol/L
I ndices of blood
1. Hemoglobin: males 130-180 g/L (13-18 g/dL)
females 120-160 g/L (12-16 g/dL)
2. Hydrogen ion: arterial blood 35-46 nmol/L (pH= 7.36-7.44) (38
o
C)
3. Oxygen (Po
2
) in arterial blood: 11-15 kPa (85-105 mm Hg)
4. Bicarbonate total (CO
2
) 22-30 mmol/L
5. Carbon dioxide (Pco
2
) in arterial blood: 4.5-6.0 kPa (35-46 mm Hg)

Osmotic function of blood. Blood maintains osmotic pressure inside the blood vessels. This func-
tion is carried out by blood proteins, chiefly albumins, and by Na
+
cations. Inside the erythrocytes, the
role of osmotic pressure regulation is assigned to hemoglobin and K
+
ions. The lowered blood plasma
concentration of proteins, or hypoproteinemia, leads to a decrease in the oncotic pressure in blood
capillaries and to oedematous disturbances. This has been observed in starvation (nutritional oedema),
in impaired hepatic albumin formation, and in other disturbed states. An increase in concentration of
107
proteins and sodium ions in blood plasma is conducive to retention of water in vascular beds; this state
is referred to as hypervolemia.
Buffer systems of blood. Since the blood is not a simple extracellular fluid, but rather a suspen-
sion of cells in a liquid medium, its acid-base equilibrium is maintained by a cooperative participation
of the buffer systems of blood plasma and blood cells, primarily erythrocites. The following buffer
systems of the blood are distinquished: plasmic systems (bicarbonate, phosphate, organic phosphate,
proteinic), and erythrocytic systems (hemoglobinic, bicarbonate, phosphate).
All buffer systems resist changes in the acid-base equilibrium. The equilibrium may be perturbed
as acidic materials are accumulated, for example, ketone bodies in diabetes mellitus. This state is cal-
led acidosis. Acidosis may be compensated, when the pH of blood remains unchanged, or uncompen-
sated, when the pH is altered. Two forms of acidosis, metabolic and gaseous, are distinquished. Meta-
bolic acidosis occurs due to retention of acidic metabolites, chiefly organic acids, in the organism. It is
accompanied by diminution of the alkali reserve of the blood, since acids displace H
2
CO
3
from bicar-
bonates. Gaseous acidosis results from accumulation of carbonic acid in the organism. In gaseous aci-
dosis, the blood alkali reserve is increased, since H
2
CO
3
makes part thereof.
Accumulation of alkaline substances in the blood is referred to as alkalosis, of which two forms,
metabolic and gaseous, are also distinquished. The former one is concomitant with an increase of the
alkali reserve in blood; the latter one develops due to an excessive expiration of carbonic acid through
the lungs, with a depletion of the alkali reserve.
Detoxifying function of blood. This function provides for detoxification and neutralization of
materials supplied to the blood. Detoxication is effected by diluting the toxicants and their binding
mainly to the albumins of blood plasma. This reduces the probability for penetration of extraneous
materials into the tissues and facilitates their expulsion from the organism. Alongside this passive neu-
tralization, toxicants can be actively detoxified by enzymes found in blood plasma and blood cells. To
exemplify, the detoxification of alcohol by alcohol dehydrogenase, of various amines by amine oxida-
ses, of curare-like compounds (succinyldicholine) by choline esterase of the blood, etc., may be quo-
ted.

LECTURE 26

Subject: RESPIRATORY FUNCTION OF THE BLOOD. HEMOSTATIC
FUNCTION OF BLOOD

In the blood, a variety of materials are transported by mechanical transport: nutrients, gases (O
2

and CO
2
), hormones, vitamins, etc.
Oxygen transport. The human organism requires an incessant supply of oxygen which is taken up
by the lungs from the ambient atmosphere to be carried to the tissues where oxygen is consumed in
reactions of aerobic oxidation of materials. In the normal state, the human tissues consume 200 to 250
ml of oxygen per minute. The human daily requirement of oxygen is of the order of 300 liters; under
heavy physical exertion, the oxygen consumption is increased by a factor of several tens. The mean
lung capacity is taken to be 6 litres, and the amount of oxygen contained in the alveolar air does not
exceed 850 ml, which is sufficient to sustein the human organism for a span of merely 4 minutes.
The blood is saturated with oxygen in the lungs. Diffusion of oxygen from the alveoli pulmonis to
the blood is effested owing to the alveolar-capillary partial oxygen pressure drop, which is: 13.83 kPa
(P
O2
in alveoli) 5.98 kPa (P
O2
in lung capilllaries) = 7.85 kPa. Oxygen that has passed through the
capillary wall dissolves in the blood plasma and then, across the erythrocytic membrane, penetrates
into the erythrocytes to become bound to hemoglobin. One gram of hemoglobin is capable of binding
1.34 ml of oxygen. Taking into account that the blood concentration of hemoglobin is 140-160 g/liter,
one will have easily estimated that one liter of blood binds a maximum of 180 to 210 ml of oxygen
(providing that all available hemoglobin is saturated with oxygen). The quantitative measure for
hemoglobin-bound oxygen in blood is called the oxygen capacity of blood. It chiefly depends on the
hemoglobin concentration in blood.
During respiration under normal atmospheric conditions, hemoglobin is never saturated complete-
ly with oxygen, but to some 95-97% (to obtain complete saturation, the O
2
percentage in the inspired
air should be equal to 35% as compared to the usual 21%). It follows therefore that under normal at-
mospheric pressure, the oxygen concentration in blood (in the form of oxyhemoglobin) is about 200
108
ml/liter. A small amount of oxygen (about 3 ml/liter) remains dissolved in blood plasma. Thus, hemo-
globin, through binding a 60-fold excess of oxygen, as compared to simple dissolution of O
2
in blood
plasma, provides for a 60 times more efficient transport of oxygen to the tissues; otherwise, the blood-
stream rate should have been increased by a factor of 50-60. For this reason, the human organism can-
not exist without hemoglobin.
Saturation of hemoglobin with oxygen depends upon the partial oxygen pressure. This relation-
ship is represented by a sigmoid curve (S-curve) whose shapr, as has been noted previously, is defined
by mutual influence of the hemoglobin subunits on their ability to bind oxygen. Owing to this relation-
ship, the oxygen supply of tissues can be ensured at small drops in the partial oxygen pressure: from
13.03 kPa in lung capillaries to 3.99-5.32 kPa in tissue capillaries.
The erythrocytes possess a special regulatory mechanism which varies the affinity of hemoglobin
to O
2
. These facilities are provided by 2,3-bisphosphoglycerate. Its low concentration in lung capilla-
ries increases the affinity of hemoglobin to O
2
and favours the formation of oxyhemoglobin, i.e. the
dissociation of oxyhemoglobin at low 2.3-bisphosphoglycerate level is suppressed. An increased pro-
duction regulatory sites of hemoglobin molecule, it makes the hemoglobin-to-O
2
affinity decrease and
facilitates the uptake of oxygen by tissues. Actually, this process is observed in the tissue capillaries.
At the arteriolar end of the blood capillary, oxygen diffuses through the capillary wall into the
intercellular medium and then enters the cells. Initially, the plasma-dissolved oxygen passes across the
erythrocyte membrane, becomes dissolved in blood plasma and transported to the cells interior. The
delivery of oxygen to peripheral tissues leads to a drop in the partial oxygen pressure at the venular
end of the capillary and to a loss in oxyhemoglobin concentration along the capillary length gives a
general idea of the oxygen requirement of tissues: the higher this oxygen concentration difference, the
greater the requirement in oxygen for a given tissue. The release of oxygen from oxyhemoglobin at the
arteriolar capillary end is also dependent on oxygen consumption in oxidative reactions within the
cells. If, because of certain reasons, the oxygen consumption in tissues becomes smaller, then, owing
to its sparing solubility in liquid medium, oxygen stops to be delivered from the arterial blood channel.
In this case, a drop in oxygen arteriolar-venular difference and a high oxyhemoglobin pressure at the
venular end are observed.













Figure 61. Scheme of HbO
2
production, curve of cooperative effect

The acidic properties of hemoglobin during oxygen transfer are liable to variations. Hemoglobin
is a weaker acid than oxyhemoglobin, and for this reason, as oxyhemoglobin is formed, the medium in
lung capillaries becomes more acidic: HHb + O
2
HbO
2
+ H
+

As the blood pH value is lowered (e.g. due to the accumulation of acidic ketone bodies in diabetes
mellitus or prolonged starvation), the saturation of hemoglobin with oxygen in the lungs is seen to
difinish. In return, the release of oxygen from oxyhemoglobin in tissues is facilitated. An increase in
pH leads to the opposite effect.
Transport of carbon dioxide (CO
2
). The transport of carbon dioxide from tissues to lungs is also
associated with hemoglobin. Carbon dioxide, as supplied to the tissue capillary blood, is consumed for
production of carbonic acid in the erythrocytes through the aid of carboanhydrase acting as a catalyst
for the reversible reaction: CO
2
+ H
2
O H
2
CO
3
. Carbonic acid dissociates into H
+
and HCO
3
-
. Ho-
wever, no acidification of the medium occurs, owing to hemoglobin which binds the H
+
ions to facili-
tate the oxyhemoglobin dissociation. HCO
3
-
anions interact with K
+
cations (whose concentration wit-
hin the erythrocytes is very high) to produce hydrocarbonates: KHCO
3
in erythrocytes, and NaHCO
3


109
in blood plasma, the Na
+
concentration in blood plasma being higher than that in erythrocytes. Owing
to the hemoglobin buffering action, carbonic acid in the tissues becomes neutralized and the blood pH
varies only within a narrow range (by less than 0.1 pH unit). Partially, carbon dioxide is bound by the
NH
2
groups of hemoglobin to produce carbohemoglobin.
It is to be inferred, therefore, that the CO
2
transport from tissues to lungs effected through the
agency of bicarbonates (KHCO
3
and NaHCO
3
) and carbhemoglobin, hemoglobin being involved,
directly or indirectly, in both processes.
In the lungs, the processes reverse to those that take place in the tissues occur. In the blood of the
lungs, the formation of oxyhemoglobin leads to a release of H
+
ions within the erythrocyte cells. The
H
+
ions released from oxyhemoglobin react with the erythrocytic KHCO
3
to yield H
2
CO
3
, and the K
+

ions become bound to oxyhemoglobin: HbO
2
+ K
+
K
+
HbO
2
.
H
2
CO
3
suffer decomposition by erythrocytic carboanhydrase, and the CO
2
formed is expired
through the lungs.
Thus, there exists a close relationship between the supply of tissues with oxygen and the dischar-
ge of carbon dioxide from them owing to the participation of hemoglobin in these two processes.
Hemostatic function of blood. Hemostasis, or arrest of bleeding to prevent blood loss, is an im-
portant function of the blood. Involved in this process are the blood coagulation system, thrombocytes,
and vascular wall.

Figure 62. Scheme of blood coagulation

The vascular traumatoses and other interferences causing stripping of the endothelium of vessels
expose the subendothelial collagen of vascular wall serving as a matrix on which a blood clot, or
thrombus, composed of blood components, is formed. The primary thrombus is formed from
thrombocytes adhering to the damaged site of vascular wall. But the primary thrombus does not arrest
bleeding it rather elicits the release of thrombocyte components that trigger off the enzymic system of
blood coagulation to form a polymeric fibrin mesh from the blood plasma fibrinogen. Blood cells
become entangled in the mesh fibres and condense to form a thrombus which stops bleeding at the
damaged spot and plugs the lumen of the vessel. The vessel patency may be restored by dissolving the
thrombus. This process is accomplished by the anticoagulative, or fibrinolytic, enzymic system. Both
systems, coagulative and fibrinolytic, are involved in monitoring the hemostasis. The former system
leads to the formation of polymeric fibrin wall, and the latter, to hydrolysis of fibrin.
Blood coagulation has two mechanisms: intrinsic and extrinsic.Blood clotting is a multistage self
accelerating process in which plasmic and thrombocytic factors are involved. To date, 13 plasmic and
11 thrombocytic factors have been described. We wish to merely point out that three major stages are
constitutive of the basis of this process. On the first stage takes place activation of prothrombokinase
110
(X factor, or Hageman factor), which participates in the formation of thrombin plasma from prothrom-
bin, and then thrombin participates in the thrombin-assisted formation of fibrin from fibrinogen.

! .

Fibrin is the building material for the thrombus. To dissolve the thrombus, plasminogen should be
activated to produce plasmin which is responsible for fibrin hydrolysis in the thrombus.






















In the organism, blood coagulation is controlled by agents that either accelerate the process (pro-
coagulants), or decelerate it (anticoagulants). Natural anticoagulants include heparin (heteropolysac-
charide), fibrinolysin, and antithrombin. Synthetic anticoagulants-antivitamins K: neodicumarin, pe-
lentan, sincumar, dicumarol, warfarin. Procoagulant-vitamin K is cofactor (coenzyme) of prothrombin
synthesis.
Regulatory, or hormonal, function of blood. The blood cells and blood plasma are sources for
production of various extracellular regulators involved in the control of metabolism and functions of
tissues and organs. These regulators belong to local hormones, or hormonoids. Basophils generate he-
parin and histamine; eosinophils histamine and serotonin; and thrombocytes serotonin. The secre-
tion of histamine and serotonin produces local changes in the capillary permeability, contractility of
vascular smooth muscles, and leads to the development of allergic reactions. Heparin as an activator
for lipoprotein lipase and as an anticoagulator takes part in the respective controls of lipids metabolism
and blood clotting.
The blood plasma proteins serve as substrates for the formation of biologically active polypepti-
des grouped under the common name kinins. They include bradykinin, kallidin and methionyl-lysyl-
bradykinin. Kinins are produced from their inactive precursors called kininogens. Kinins are released
from the blood plasma kininogens by the action of proteolytic enzymes called kininogenases. These
include kallikreins of blood plasma and tissue, derived from inactive enzymes kallikreinogens. The tis-
sue kallikreinogens are activated by katepsins or trypsin in the pancreas, and the blood plasma kalli-
kreinogens, by blood plasma proteinases Hageman factors (blood coagulation system enzyme), plas-
min (fibrinolytic enzyme), and trypsin (supplied to blood plasma, especially in affected pancreas).
The physiological role of kinins consists in the control of the bloodstream rate, blood pressure,
and capillary permeability. Kinins produce dilatation of peripheral citculatory and coronary vessels,
decrease arterial pressure, increase capillary permeability, and stimulate the cardiac cycle. In addition,
kinins are capable of eliciting the contraction of smoth muscles of nonvascular organs (bronchi, uterus,
and intestine); they stimulate intracranial baro- and algesic receptors and perturb the intracranial pres-
sure of the cerebrospinal fluid.

111
Under physiological conditions, the systems for production and inactivation of kinins are equili-
brated. Pathologic alterations occur due to an excessive kinin generation, which is concomitant with
the development of local inflammations and impaired blood circulation.
Blood as source for medicinal preparations. The blood is used as a raw material for producing a
variety of medicinal preparations which, by their therapeutic applications, are divided into four
groups:
- systemic effect agents (albumin, protein, native blood plasma)
- immunologically active preparations (gamma-globulin, antistaphylococcic, interferon)
- hemostatic preaparations (antihemophilic plasma, thrombin, fibrin sponge, fibrin film, fibrino-
gen)
- antianemic and stimulating preparations (polyobolin dry powdered protein components of
blood plasma, eryheme-dehydrated hemolyzate of erythrocytes, etc.).

LECTURE 27

Subject: FUNCTIONAL BIOCHEMISTRY OF THE LIVER

The liver takes a central place in the organism metabolism. Specific organization of the enzymic
hepatic apparatus and its anatomic routes to other organs enable the liver to participate practically in
all types of metabolism and to maintain at appropriate concentration levels many vitally important blo-
od components in the organism. One may safely assert that the functional specialization of the liver
exemplifies a specific biochemical altruism, i.e. provision of essential conditions for normal functio-
ning of other organs and tissues in the organism. This bridge forward an explanation of the specificity
of biochemical hepatic processes which, on the one hand, are oriented to the production of various
compounds for other organs and, on the other hand, provide defense for these organs from toxicants
formed therein, or from extraneous compounds invading the organism.
The liver is involved in the following biochemical functions:
1. regulatory-homeostatic
2. ureapoietic
3. biligenic
4. excretory
5. detoxifying.
Regulatory-homeostatic function. The liver participates in the metabolic control of nutrients:
carbohydrates, lipids, proteins, vitamins, and, in part, water-mineral compounds as well as in the meta-
bolism of pigments and nitrogenous nonproteinic materials.
The carbohydrate metabolism control effected owing to the fact that the liver is actually the only
organ capable of maintaining a constant glucose level of the blood, even under starvation conditions.
Glucose produced by the liver in the course of glycogenolysis and gluconeogenesis, is supplied to the
blood to be consumed primarily by the nervous tissue or, when its supply from the intestine is excessi-
ve, to be stored as glycogen (see Carbohydrate metabolism).
The lipid metabolism control effected through a variety of liver-biosynthesized lipids (choleste-
rol, triacylglycerides, phosphoglycerides, etc.) which are secreted into blood to be delivered to other
tissues. The amount of cholesterol produced by the liver is larger than that ingested in food: in
humans, the daily dietary intake of cholesterol is about 0.3-0.5 g, while the cholesterol that is daily
produced in the liver amounts to 2-4 g,. The distribution of lipids over the organs and tissues is accom-
plished by the liver. Apoproteins of o- and |-lipoproteins are formed in the liver. In addition, fatty
acids are catabolized in the liver to yield ketone bodies which are used as an energy source for extra-
hepatic tissues.
The protein metabolism control is effected by the liver owing to the intense synthesis of proteins
and oxidation of amino acids therein. The human organism synthesized about 80-100 g of proteins per
day of which a half is accounted for by the liver. Unlike other organs and tissues a major portion of
proteins synthesized by the liver (mainly albumin) is supplied for consumption by other organs. The
liver produces daily about 12 g of blood plasma albumins and most o- and |-globulins of the blood. In
addition, the liver is a producer of other blood plasma proteins involved in the coagulative and anti-
coagulative blood systems), choline esterase, transport proteins (ferritin, ceruloplasmin).
112
In the liver amino acid metabolism is the most active process, viz. synthesis of nonessential ami-
no acids, of nonproteinic nitrogenous compounds from amino acids (creatinine, glutathione, nicotinic
acid, purines and pyrimidines, dipeptides), oxidation of amino acids with a release of ammonia.
During starvation, the liver is the first to loose its reserve proteins in supplying other tissues with ami-
no acids. The protein losses in the liver amount to about 20%, while in other organs they do not exce-
ed 4%.
The involvement of the liver in vitamin metabolism is associated with deposition of vitamins in
it, mostly fat-soluble ones, synthesis of certain vitamins (nicotinic acid) and coenzymes, and conver-
sion of calciferols to 25-hydroxycalciferols.
The involvement of the liver in water-mineral metabolism consists in that the liver supplements
the function of the kidneys in maintaining the water-salt equilibrium and acts as a kind of an internal
buffer for the organism. The available evidence indicates that the liver retains Na
+
, K
+
, Cl
-
. Ca
2+
ions
and water and secretes them into the blood. Besides, the liver stores microelements (iron, copper) and
participates in their distribution among other tissues through the agency of transport proteins.
The involvement of the liver in the metabolism of nitrogenous bases of nucleic acids shows up
in the synthesis of these species from simpler compounds and their subsequent oxidation to uric acid.
Nitrogenous bases are utilized by other organs for the synthesis of nucleosides, nucleotides, and
nucleic acids, uric acid being the end metabolite.
Ureapoietic function. Liver is the only organ in possession of all the enzymes involved in the
cycle for production of urea from ammonia. The ammonia generated within other tissues becomes
converted in the liver to a neutral product (urea), which is excreted into blood. An intense catabolism
of proteins and nonproteinic nitrogenous compounds (amino acids, purines, pyrimidines, and biogenic
amines) is accompanied by enhanced hepatic ureapoiesis, increased blood concentration of urea and
increased urinary discharge of urea.
Biligenic and excretory functions. Liver produces a special liquid excretion, bile, which is relea-
sed in the small intestine. Liver is the only producer of bile acids and their conjugates, which are used
in digestion and absorption of lipids in the intestine. The bile is composed of bile acids, proteins (albu-
mins and globulins), cholesterol and its esters, mineral compounds (Ca
2+
, Na
+
, K
+
), water, pigment
metabolites (bilirubin glucuronides), inactive products of hormone and vitamin metabolism, extra-
neous invaders in the organism. A disorder of the excretory hepatic function disfavours the digestion
and uptake of lipids and leads to an accumulation of toxic metabolites derived from pigments and
foreign substances.
Detoxifying function. Liver is the major organ involved in the detoxification and neutralization
of intrinsic metabolites and extraneous substances. The participation of the liver in pigment metabo-
lism is manifested by conversion of chromoproteins to bilirubin in the cells of reticuloendothelial sys-
tem (RES) present in the liver, by conjugation of bilirubin in the liver cells, and by hepatic breakdown
of intestinal urobilinogen to nonpigmentary products.
Disturbances of liver functons. Damages inflicted on the liver by infections agents or chemicals
disturb its functions, all or some of them. A change in the concentration of substances secreted into the
blood from the liver can indicate such disturbances. These substances are glucose, cholesterol, phos-
pholipids, vitamins and 25-hydroxycalciferol, uric acid, urea, etc.
Disturbances in the regulatory-homeostatic function are distinctly visible in compositional altera-
tions of the blood plasma proteins; the relevant symptoms are:
- dysproteinemia, i.e. disbalanced ratio of plasmic protein fractions (albumin/globulin quotient is
low);
- reduced concentrations of albumin, fibrinogen, prothrombin, and other proteinic clotting fac-
tors, transport proteins (o- and |-lipoproteins, transferrin, ceruloplasmin, hormone- and vitamin-tran-
sporting proteins);
- reduced activity of choline esterase, and some others.
In hepatic pathology, the disturbances of ureapoiesis are manifested in a reduced blood concen-
tration of urea and in a decreased daily urinary content of urea, while the disordered detoxifying func-
tion shows up in reduced rates of conversion and conjugation of metabolites (e.g. bilirubin, indole,
scatole) and foreign compounds (benzoic acid, arylamines, alcohols) supplied in food or administered
to the organism.
The inhibition of bile acid production manifests itself in reduced concentrations of bile acids in
the bile, and in symptoms of disordered digestion and uptake of lipids. The impaired excretory func-
tion of the liver leads to the retention in the organism of substances normally excreted in the bile (bile
113
acids, bilirubin glucuronides, various drugs and toxins). Their increased concentrations in the blood
and urine are diagnostic of alterations in the excretory function of the liver.

LECTURE 28

Subject: FUNCTIONAL BIOCHEMISTRY OF THE KIDNEY

Specific functions performed by the kidney are the following:
1. uropoietic and excretory
2. regulatory-homeostatic
3. detoxifying
4. incretory (endocrine).
The major, vitally important function of the kidney is uropoiesis (production of the urine), which
is followed by the excretion of substances, including extraneous ones that enter the organism.
The functional unit of the kidney is the nephron. The uropoiesis in the nephrons is effected via
ultrafiltration of blood plasma at the glomerules, via tubular reabsorption of materials (in tubules and
collecting ducts), via secretion in the urine of certain excrets (in tubules). About 180 liters of blood
plasma ultrafiltrate (primary urine) is filtrated by the kidney per day, at a rate of 120 ml/min. Over
99% of the total of ultrafiltrate is returned to the extracellular fluid by reabsorption. Urine discharged
from the organism amounts to 1.0-2.0 liters per day.
All compounds contained in the primary urine are differentiated into threshold and no-threshold
substances. Normally, the former are reabsorbed completely and are excreted into the urine only when
their concentration in the primary urine exceeds a certain threshold value. The latter substances are not
reabsorbed and are excreted in the urine in proportion to their concentration in the blood plasma. The
reabsorption proceeds either by simple diffusion, or by active transport. Most materials are reabsorbed
by active transport, which requires large energy expenditure. For this reason, the active transport
system in the renal tubules is highly developed: the activity of Na
+
, K
+
-ATPase which provides for the
Na
+
, K
+
gradient in the secondary active transport, and the activity of protein carrier systems subser-
ving various substances are quite efficient. The kidneys are rich in mitochondria, and differ in oxygen
consumption. This enables the kidneys to produce much energy in the course of oxidative phosphory-
lation. In the renal generation of energy, glucose, fatty acids, acetone bodies, and amino acids are utili-
zed as energy sources.
The primary urine is produced by ultrafiltration of blood through the pores of the glomerular
basal membrane whose size is about 4 nm. The ultrafiltrate contains all the blood plasma components,
except the proteins with a molecular mass above 50,000.
The uropoietic function of the kidney is closely related to the renal capacity to control osmotic
pressure, salt-and-water balance, and acid-base equilibrium in extracellular fluids, blood included, of
the organism.
The kidney regulates the acid-baseequilibrium in the blood and favours the urinary excretion of
acidic substances and the retention of the alkali reserves (bicarbonates) in the organism. In the course
of metabolism, acidic compounds (lactate, ketone bodies, and carbonic acid) are chiefly formed. The
removal of volatile acidic substances is effected by pulmonary expiration, and of nonvolatile substan-
ces, by renal filtration. Acid anions are mainly neutralized by sodium cations and, because of this, are
excreted in the urine as sodium salts (Na
2
HPO
4
, NaHCO
3
, NaCl, and sodium salts of organic acids).
To preserve hydrocarbonate HCO
3
-
(which is the alkali reserve of the blood), Na
+
ions are reabsorbed
in the distal tubules and the replaced in the urinary salts by H
+
and NH
4
+
that are produced by the tubu-
lar epithelium. The reabsorption of Na
+
is followed by reabsorption of HCO
3
-
anions these remain in
the organism. Salts of greater acidity (NaH
2
PO
4
and NH
4
Cl) and acids (carbonic, lactic, acetylacetic,
and |-hydroxybutiric) are excreted in the urine. The urine test becomes distinctly acidic, and the pH
may be as low as 4.5 while the alkali reserve of the blood is preserved.
In the kidney, the detoxication of foreign compounds is accomplished by binding the toxicants to
glycine, acetic and glucuronic acids. Moreover, the renal oxidation of certain alcohols and other mate-
rials also occurs. In the cells of renal connective tissue, extracellular regulators of hormonoid type (for
example, prostaglandins) are formed. The kidney is also involved in the hormonal control of vascular
tension and pressure.
114

Characterization of urine components in norm and in pathology
The urinary excretion of various materials reflects alterations in the processes that occur in the
kidney and other tissues and organs of the organism. The daily volume of final urine amounts to 1.0-
2.0 liters and the dry weight of final urine is about 60 g. Since the urine is a filtrate of blood plasma, it
appears expedient to consider the urinary concentrations of various groups of biological materials
from the standpoint of their occurrence in the blood plasma.
Proteins. In norm, the daily urinary excretion of proteins amounts to about 30 mg, which is not
detectable by common laboratory techniques and routinely specified as traces, or absence of urinary
proteins. Among the urinary proteins, enzymes are also present. The origin of normal urinary pro-
teins is different.
In pathology, the urinary protein concentration may be increased; depending on the location of
the damage, prevalent in the urine may be either plasmic proteins, or cellular proteins of the urinary
tract. In inflammatory renal diseases (glomerulonephritides), the permeability of the basal membrane
of nephron glomerulus increases; proteins are filtered in an amount above normal and fail to be reab-
sorbed completely. Disturbances in the tubular protein reabsorption (nephroses) are conducive to a
similar pathology. For this reason, in patients with glomerulonephritides and nephroses the urinary ex-
cretion of proteins may vary from 1 to 15-40 g per day. Nonetheless, even in such a contingency, the
urinary proteins concentrations are small and can be detected only using special techniques. For exam-
ple, in pancreatites, an enhanced activity of o-amylase and trypsin is observed both in blood and urine.

Nonproteinic nitrogenous urinary components
Urea is a major nitrogenous component of the urine. The normal excretion of urea is 333 to 583
mmol per day, which accounts for 60% to 80% of the overall urinary nitrogen. An increased urinary
concentration of urea is observed in the states with pronounced catabolism of proteins and other nitro-
genous components (starvation, burns, traumatism, atrophy of tissues, etc.). A decreased excretion of
urea is observed in affected liver (urea-producing organ) and in impaired in the blood (this state is cal-
led azotemia).
Uric acid. Normally, the urinary excretion of uric acid is 2.35 to 5.90 mmol per day. Its increased
urinary concentration is observed in a diet rich in nucleic acids or as produced by breakdown of cells
and tissues, for example, leucocytes in patients with leucosis.
Creatinine. In norm, the urinary excretion of creatinine is 4.4 to 17.6 mmol per day; variations in
creatinine concentration are dependent on muscular development. Physiological excretion of creatini-
ne is normal only in children. In adult humans, creatinuria is a sign of pathology (e.g. muscular dystro-
phy).
Amino acids. In norm, the urinary excretion of amino acids is 0.29 to 5.35 mmol per day (as ba-
sed on nitrogen). The urinary concentrations of glycine, histidine, and alanine are higher than those of
other amino acids. In pathology (e.g. burns, diabetes mellitus, affected liver, and muscular dystrophy)
hyperaminoaciduria may occur. Heriditary hyperaminoaciduria is associated with defective proteins-
carriers for amino acids in the proximal renal tubules. In a disordered amino acid tissue metabolism,
the urinary excretion of normally nonexcretable amino acid metabolites occurs (e.g. homogentisine
acid, in alcaptonuria; phenylpyruvic acid in phenylketonuria).
Ammonium salts. In norm, the urinary excretion of ammonia as a component of ammonium salts
(ammonium chloride) is 30-60 mmol per day. In pathology, an increased urinary elimination of ammo-
nium salts may be observed (in diseases accompanied by acidosis). A diminished excretion of ammo-
nium salts occurs in diseases associated with alkalosis, in renal diseases due to affected distal tubules
in which ammoniogenesis takes place.
Hippuric acid. The urinary excretion of hippuric acid is dependent solely on the amount of inges-
ted vegetable food, since in the organism this acid endogenically is not produced. Commonly, the dai-
ly urine contains to 5.5 mmol of hippuric acid.
I ndican (indoxyl sulphuric acid). Normal urine contains indican (in the form of potassium indo-
xyl sulphate) in trace amounts. In detectable quantities, indican appears in the urine on excessive ali-
mentary intake of meat products; it also occurs as a byproduct of putrefactive processes in the intesti-
ne.
Nitrogenous pigments. Representative of these is stercobilinogen, a product of hemoprotein brea-
kdown. Stercobilinogen is convertible to stercobilin and normally is excreted in the urine. In patholo-
gy, urinary excretions contain bile acids and a variety of bile pigments, for example, in affected liver
and in toxicoses conducive to hemolysis.
115

Nitrogen-free components of urine
Glucose and other monosaccharides. In norm, the daily urine contains a more 0.3-1.1 mmol of
glucose. Such amounts escape detection by conventional analytical techniques; for this reason, glucose
is not reckoned as a component of normal urine. However, in excessive dietary intake of carbohydra-
tes, when the glucose concentration in blood attains a threshold value, i.e. of the order of 8.3-8.8
mmol/liter, alimentary glucosuria may develop in the organism. In pathology, glucosuria occurs due
either to an increased blood glucose concentration, or to a defective carrier protein involved in glucose
reabsorption in the renal proximal tubules. The former case is the most commonly encountered in the
clinic, for example, in diabetes mellitus or in steroid diabetes. The latter case is the so-called renal dia-
betes. For example, the occurrence of fructose or pentose in the urine (renal fructosuria or renal pento-
suria) is indicative of affected transport systems of the renal tubules.
Lactic and pyruvic acids. In norm, the daily urinary excretions of lactic and pyruvic acids amount
to 1.1 and 0.11 mmol, respectively. An increased concentration of lactic acid in the urine is observed
under intensive muscular work and in hypoxia. An increased urinary excretion of pyruvic acid occurs
in diabetes mellitus and in B
1
hypovitaminosis.
Ketone bodies. In norm, the daily urine contains 20 to 50 mg of ketone bodies. At this level, they
are not detectable by the analytical methods currently employed in the clinic. In pathology, increased
concentrations of ketone bodies, i.e. a state called ketonuria, occur in diabetes mellitus, steroid diabe-
tes, and starvation.
Mineral salts. In norm, the daily urine contains (in mmol): sodium, 174-222; potassium, 61-79;
calcium, 4.02-4.99; inorganic phosphorus, 33. In pathology, an increase in urinary excretion of so-
dium and a decrease in excretion of potassium are observed in the adrenal hypofunction; the reverse
situation occurs in hyperaldosteronemia and when mineralocorticoids and glucocorticoids are prescri-
bed as drugs. A decreased urinary concentration of calcium and a distinct phosphaturia are observed
when large doses of vitamin D and parathyrin are administered; a high urinary loss of calcium is cha-
racteristic of rickets and hypoparathyroidism.

LECTURE 29

Subject: BIOTRANSFORMATION OF DRUGS AND POISONS

All the substances supplied to the organism in a variety of ways pass through several basically
similar stages such as absorption, distribution (mechanical transport), and excretion. The transit rate of
substance at these stages may either be increased, or lowered, depending on the structural features and
physico-chemical properties of a substance as well as on its affinity to biological molecules. The disci-
pline, dealing with rate characteristics at the stages in which any substance entering the organism is in-
volved, is referred to as chemobiokinetics which treats, in a broader sense, movements of substances
in the living organism. Conceptually, chemobiokinetics is divided into three subdisciplines: pharma-
cokinetics, toxicokinetics, and biokinetics. Pharmacokinetics confines itself to the study of drugs; toxi-
cokinetics, to the study of toxic substances; and biokinetics, to the study of substances not alien to the
organism. In many respects, this classification is rather arbitrary, since the distinction between a drug
and a poison in many instances may be evasive. Moreover, even autobiogenous compounds taken in
improper doses may exhibit toxic properties. The subsequent history of a substance after its uptake by
the organism is dependent to a significant degree on the rates at which it is converted by various enzy-
mes, i.e. on its metabolic transformations. In point of fact, the metabolism of biogenous substances
and xenobiotics used as drugs is governed by the laws of enzymic kinetics. Biogenous substances,
being natural substrates for enzymes, are converted at the rates characteristic of catalytic properties of
the enzymes involved. The metabolic evolution of xenobiotics is dependent on the occurrence of enzy-
mes capable of catalyzing the conversion of these xenobiotics. If no enzymes that are potentially capa-
ble of catalytic intervention of the xenobiotics are available, the xenobiotics behave as metabolically
inert. Apparently, in the course of evolution, highly substrate-specific enzymes have laid a basis for
the intrinsic metabolism in living organisms, while the enzymes with low specificity towards substra-
tes have taken up defense functions aimed at the inactivation of extraneous invaders.
116
Biochemistry studies enzyme-assisted conversions of drugs in the organism by making use of
appropriate methods and techniques. The drug metabolism in the organism may be represented within
the framework of a general scheme:

Drug Enzymes Metabolites End Metabolites
absorption intrinsic metabolism excretion or storage

The drug metabolism is studied by determining the drugs and their metabolites in biological
fluids, tissues, and excretions as well as by estimating the activity and kinetics of enzymes involved in
the drug metabolism.
Experimentally, the two approaches are used in the studies on metabolism of xenobiotics. In the
clinic, the drug metabolism is assessed, as a rule, by measuring the concentration of administered drug
and its metabolites in blood, urine, and other excretions.

Stages in the metabolism of xenobiotics
Biogenous substances, as distinct from xenobiotics, are involved in the conventional metabolic
process. Xenobiotics, in the course of their conversion, are subject to two major stages: modification
(nonsynthetic stage) and conjugation (synthetic stage).
The modification stage is an enzyme-assisted modification of the initial structure of a xenobiotic
resulting either in a cleavage of bonds within the xenobiotic molecule, or in the insertion of additional
functional groups (e.g. hydroxyl or amino groups) into its molecule, or in a release of its functional
groups blocked in the initial structure (for example, by hydrolysis of ester or peptide bonds). The mo-
dification leads to an increased solubility of the xenobiotic (xenobiotic becomes more hydrophilic).
Additional functional groups are needed to enable the xenobiotic to enter the conjugation stage.
Conjugation stage is viewed as an enzyme-assisted process for building covalent bonds between
the xenobiotic and biomolecules occurring in the organisms media (e.g. glucuronic acid, sulphates,
and others). The conjugation stage terminates in the synthesis of a novel compound whose constituents
are, on the one hand, the xenobiotic moiety and, on the other hand, a conjugate (biomolecule).
Relationship between metabolism of xenobiotics and their structure Xenobiotics invaded into the
organism are liable to a chain of modifications, or nonsynthetic conversions (oxidation-reduction,
isomerization, cyclization, ring opening, and hydrolysis) carried out by the respective
enzymes (oxidoreductases, isomerases, lyases, and hydrolases):
RH + O
2
+ 2H
+
+ 2e

ROH + H
2
O
Depending on the number of functional groups in the molecule of modified xenobiotic, its conju-
gation can proceed by a variety of routes in which each of the xenobiotic functional groups becomes
bound with a conjugating agent. If the xenobiotic is not functionalized (e.g. benzene), it cannot enter
the conjugation stage. In contrast, if the introduced xenobiotic is in possession of an appropriate func-
tional group (e.g., 4-aminobiphenyl); it may become immediately engaged in the conjugation stage
with UDP-glucuronic acid.
The knowledge of principles that govern the enzyme-assisted conversions of xenobiotics provides
an opportunity to prognosticate metabolic behaviour of any xenobiotic taking into account its structu-
ral specificities.



Xenobiotic routes in the organism
Xenobiotics are either eliminated from the organism, or become accumulated in tissues. Xenobio-
tics are excreted as:
a) supplied (unmodified by enzymes);
b) metabolites (modified by enzymes);
c) conjugates (by action of conjugating enzymes);
117
d) complexed with biomolecules (for example, metal-containing xenobiotics become bound to
cysteine by glutathione and excreted as complexes).
The xenobiotics that accumulate in the organism are those capable of interacting with macromole-
cules (proteins, nucleic acids, and lipid entities). For example, organochloric compounds, which are
readily soluble in lipids, are quite resistant to catabolic conversion and are difficult to eliminate from
the organism. They tend to accumulate in lipid-rich tissues. Heavy metals (mercury, cadmium, silver,
arsenic, and lead) and preparations containing organometallic compounds become bound with proteins
and likewise accumulate in the organism.

Metabolism and physiological action of drugs
Substances introduced into organism may exhibit either medicinal or toxic properties. Common-
ly, any drug can exert both medicating and side (toxic) effects. Therefore, generally speaking, the mo-
re active the drug, the faster its toxic properties become manifest. During metabolism, the specific ac-
tivity and toxicity of xenobiotics are susceptible to alterations.
Biological activity alterations show up in:
a) deactivation, i.e. a loss of medicinal or biological activity of drugs;
b) activation, i.e. induced activity of an inactive preparation;
c) modification of the major effect, i.e. when the administered drug, on having metabolized, exhi-
bits properties different from those of the initial preparation.
The alterations of toxicity are manifested in:
a) deintoxication, i.e. a loss or reduced toxicity of drug;
b) toxification, i.e. enhanced toxicity of drug.
The above instances may be exemplified as follows:
Deactivation is observed as the functional groups responsible for the biological activity of a drug
are either eliminated from, or blocked in the drug molecule. For example, the active sulphanilamide,
after its conjugation with acetyl-ScoA, is converted to an inactive acetylsulphanilamide.
Activation is observed when the biologically active groups that have been blocked in the initial
preparation become deblocked during metabolism:

Phthalsulphathiazole hydrolysisSulphathiazole + Phthalic acid
inactive drug in organism active drug

or acquire functional groups that are necessary for eliciting the drug activity:

Benzopyrene hydrolysis in organism Hydroxybenzopyrene
inactive carcinogen,i.e.tumor-
procarcinogen producing agent

Modification of the major drug effect manifests itself as a variant of activation. For example,
codeine (morphine 3-methyl ether) exhibits mainly antitussive and mildly analgesic action. When
codeine undergoes demethylation in the organism, it converts to morphine, which is a strong analgetic.
Deintoxication resembles deactivation and is a defense reaction to the toxic effect of a drug. For
example, phenol is a toxic compound, while phenol sulphate, which is a product of phenol conjugation
in the organism, is nontoxic.
Toxication shows up as an enhanced side effect due to a drug administered into the organism. By
mechanism, toxification resembles activation. Occasionally, toxification is produced by lethal mole-
cules synthesized from the introduced compounds during their metabolism in the organism. The lethal
synthesis with the involvement of a xenobiotic leads to a metabolic block and to the death of orga-
nism. For example, the administered fluoroacetate enters the Krebs cycle in tissues to produce a toxic
product, fluorocitrate, which blocks aconitate hydratase and interrupts conversion steps in the Krebs
cycle. Toxification effects are taken into account in the development of chemicals against rodents and
other vermin.

Localization of drug metabolism in the organism
Depending on the site of conversion of biogenous preparations and xenobiotics in the organism,
the drug metabolism is classified into cavitary (enteral), extracellular (humoral), and cellular, or tissue,
types of metabolism.
The cavity, or enteral, drug metabolism is effected by hydrolytic enzymes supplied to the cavity
of gastrointestinal tract. Hydrolysis of biogenous preparations occurs with the involvement of pancrea-
118
tic and intestinal digestive enzymes. Xenobiotics whose molecules contain peptide, carboxyester, gly-
coside, amide and phosphamide bonds are also liable to hydrolysis. This process involves proteolytic
and lipolytic enzymes as well as enzymes capable of hydrolyzing glycoside bonds. In addition, a large
group of esterases (e.g. carboxyesterases and phosphatases) and phosphamidases (involved in hydroly-
sis of phosphamide bonds in drugs) are found in the intestine. Trypsin, while being a proteolytic, exhi-
bits also an esterase activity and is capable of hydrolyzing the ester bonds in xenobiotics.
Extracellular, or humoral, drug metabolism takes place in the extracellular fluids (after uptake
and subsequent circulation of a drug in the organism), i.e. in the blood, lymph, cerebrospinal, and ex-
tracellular proper, fluids. Possibly, metabolic conversions therein are chiefly confined to hydrolysis of
the preparations delivered (both biogenous and xenogenous). In the blood and other fluids, this func-
tion is performed by proteinases and esterases (e.g. pseudocholine esterase, phosphatases). In the ex-
tracellular fluids, other enzymes, for example, alcohol dehydrogenase, aminooxidases, etc., are availa-
ble in small amounts, but the activity of these enzymes is rather low. The contribution of the humoral
metabolic link to the overall drug metabolism is insignificant. At the humoral level, drug hydrolysis
plays a role in drug inactivation; this metabolic link should be taken into account.
Cellular (tissue) drug metabolism. In the cells, the whole varieties of metabolic transformations,
including those of xenobiotics, are being accomplished. However, the substances, before being subjec-
ted to the action of enzymic systems, should be transported from the site of their introduction to the
cells and allowed to penetrate the intracellular space through the cell membrane. Xenobiotics are
transported by the same mechanisms as biogenous substances. In the blood plasma, they either become
dissolved in the liquid medium, or adsorbed, mostly on albumin. In a dissolved or in a protein-bound
state, xenobiotics are delivered to the cells (tissues). They gain access to the cells mostly by simple
and facilitated diffusion; large molecules enter the cells by endocytosis. Xenobiotics synthetically
derived from biogenous substances can be actively transported across the cell membranes using natu-
ral substance transport systems.
Not all the tissues and organs are equally active when they convert xenobiotics. The most actively
engaged organ is liver which is in possession of enzymes that perform modification and conjugation of
drugs. The other organs and tissues are less active in the metabolism of xenobiotics.
The metabolic conversion of xenobiotics occurs in various organelles of the liver cells. The most
powerful metabolic system is found in endoplasmic reticulum (in microsomes). The microsomes are
fragments of endoplasmic reticulum that are formed, for example, on trituration of a tissue sample and
spontaneously close into small bladder-like structures (vesicles). Thus, with reference on its localiza-
tion, the metabolism of xenobiotics is differentiated into microsomal and extramicrosomal. The extra-
microsomal metabolism occurs in hyaloplasm, lysosomes, peroxisomes, and mitochondria.
The enzymic reactions conducive to conversion of xenobiotics may be divided into the following
major groups:
1. oxidation-reduction reactions;
2. hydrolytic reactions
3. synthetic reactions, or conjugation reactions;
4. other reactions (isomerization, ring opening, etc., which are effected by isomerases and lyases).
Microsomal oxidation of substances
In the microsomes, there are found enzymic chains for oxidation of substances. These chains are
represented by two short electron-proton transfer chains built into the membranes of endoplasmic reti-
culum or into microsomal membranes. Microsomal oxidation is connected with these chains. One of
these chains is a monooxigenase oxidation chain (in which the source of electrons and protons is redu-
ced NADP), and the other is a reductase oxidation chain, with reduced NAD as a suplier of electrons
and protons. The source of NADPH
+
in the monooxigenase chain is the pentose phosphate cycle, and
the source of NADH
+
is glycolysis.
The microsomal NADPH
+
-dependent monooxigenase chain is composed of flavoprotein (FP
2
),
with FAD for a coenzyme, and cytochrome P
450
. Flavoprotein exhibits a NADPH
+
-dehydrogenase
activity, FAD acting as an acceptor for two protons and two electrons. From flavoproteins, electrons
are transported onto cytochrome P
450
, and protons are lost into the environment (cytosol). Cytochrome
P
450
is the terminal self-oxidizable link of this chain. Like all the cytochromes, it belongs to hemopro-
teins. Its protein moiety is represented by a single polypeptide chain. The molecular mass of cytochro-
me P
450
is about 50,000. The P
450
is capable of complexing with carbon monoxide, CO. The light ab-
sorption maximum for these complexes is at 450 nm; hence the name for the given cytochrome.
119
Cytochrome P
450
performs a dual function: it activates molecular oxygen by transferring electrons
onto it, and uses the activated oxygen to oxidize substances R, with the concomitant formation of
water. Consequently, one oxygen atom adds to the oxidizable substance (RO), and the other, by
accepting two H
+
ions from the medium, makes up water.
The NADH
+
-dependent reductase oxidation chain occurs not only in the microsomal membra-
nes; it is also available in the outer mitochondrial membrane, in the nuclear membrane, and in the ery-
throcytic cell membranes. The reductase chain is thus included among the most rapid reactions of bio-
logical oxidation, but its function in the cell remains still unclear. The self-oxidizable component of
this chain capable of activating the oxygen has never been identified either; quite probable that this
function is exercised by cytochrome P
450
itself. The NADPH
+
- and NADH
+
-dependent chains can ex-
change electrons. For example, the electrons from FP
2
and cytochrome b
5
may be transferred onto
cytochrome P
450
to be used in the oxidation of substrates.

Cojugation of xenobiotics, its mechanism and role
The conjugation stage, or synthetic stage, is essential for the formation of nontoxic and easily ex-
cretable drug metabolites. By their mechanism, the conjugation reactions are divided into two groups:
- Reactions of I type. Initially, conjugating agents, i.e. biomolecules, are activated and then
transferred onto xenobiotics to form conjugates. This type of conjugation reactions occurs in all tissues
of the organism.
- Reactions of I I type. Initially, a xenobiotic is activated to be transferred onto a conjugating
biomolecule to form a conjugate. This conjugation type is of rare occurrence and is only observed in
liver and kidney.
Various groups for conjugation reactions of I and II types are distinguished, depending on the na-
ture of a conjugating species involved. In the I type reactions, glucuronide, sulphate, acetyl, methyl,
thiosulphate conjugations are to be noted, and in the II type, glycine and glutamine conjugations.
Glucuronide conjugation. UDP-glucuronic acid is the source for glucuronic acid residues in this
process. Endogenous substances and xenobiotics are subject to glucuronide conjugation (known are
glucuronides of bilirubin, steroid hormones, vitamin D, etc.). Xenobiotics can enter glucuronide conju-
gation if they possess or have acquired, during modification, a hydroxyl, carboxyl, and amino group
(commonly, in the aromatic ring), or, at least, a SH-group. The conjugation reaction proceeds with the
participation of UDP-glucuronosyltransferase by the scheme:


RXH + UDP~C
6
O
9
O
6
UDP + RX-C
6
H
9
O
6

xenobiotic UDP-glucuronic glucuronide
acid of xenobiotic

Among xenobiotics (drugs and poisons), susceptible to glucuronide conjugation are phenols, po-
lyphenols, phenolic steroids, aromatic amino acids, and others.
Sulphate conjugation. Active form of a conjugating agent is 3

-phosphoadenosine-5

-phospho-
sulphate (PAPS for short). PAPS, which may also be designated as PAP~SO
3
H, is a source of labile
sulphate groups used in the conjugation of natural compounds and xenobiotics. The natural substances
as subject to sulphate conjuation include endogenous toxic products, e.g. indole, scatol, phenol as well
as steroids, iodothyronines, tocopherols, and others.
The sulphate conjugation reaction proceeds with the involvement of a special enzyme, sulpho-
transferase, according to the scheme:

RXH + PAP~SO
3
H RX-SO
3
H + PAP
xenobiotic PAPS sulphate 3-phosphoadenosine-
of xenobiotic 5-sulphate
Acetyl conjugation. The source of labile acetyl groups in this variety of conjugation reactions is
acetyl~ScoA, which is produced by degradation of carbohydrates, triacylglycerides, and amino acids.
Endogenous substances and xenobiotics containing a free NH
2
group may be acetylated.
N-acetylation is an essential biochemical reaction in the synthesis of monosaccharide derivatives
(N-acetylglucosamine, N-acetylgalactosamine, and neuramic acid) that are further used in the synthe-
sis of heteropolysaccharides. N-acetylation is also a route to neutralization of biogenous amines: sero-
tonine, histamine, GABA, and others. N-acetylation of histones and nonhistonic chromatin proteins is
an important regulatory mechanism of DNA transcription. For endogenous substances, the only case
of O-acetylation has been reported, which is a reaction of acetylcholine formation.
120
Xenobiotics possessing a free NH
2
group (commonly, on the aromatic ring) are subject to acetyla-
tion. This reaction is effected by means of a special acetyltransferase called arylamine-N-acetyltrans-
ferase. This enzyme exhibits a low specificity to xenobiotics to be acetylated. The reaction proceeds
by the scheme:

RNH
2
+ CH
3
-CO~SCoA R-NH-CO-CH
3
+ CoASH

Among the xenobiotics susceptible to acetylation, sulphanylamides, isonicotinic acid hydrazides,
and aniline derivatives can be mentioned; these preparations are widely used in medical practice.
Methyl conjugation. In this reaction methyl groups derived from the active form of methionine,
S-adenosylmethionine, serve as a conjugating agent. S-adenosylmethionine is a participant in nume-
rous reactions of methylation of endogenous compounds. It is also a methyl group donor for conjuga-
tion reactions of xenobiotics (RXH), which proceed with the involvement of methyltransferases, ac-
cording to the scheme:

RXH + S~Adenosylmethyonine RX-CH
3
+ S~Adenosylhomocysteine

Xenobiotics containing an NH
2
group or heterocyclic nitrogen, as well as OH and SH groups, are
subject to methylation by addition of methyl groups to N, O, and S atoms. Among the preparations
used in therapy, liable to methylation are mono- and polyphenols, and heterocyclic compounds of
pyridine, quinoline, isoquinoline, and thiouracil type.
Thiosulphate conjugation. This kind of conjugation is used in the enzymic detoxication of cyani-
des. The transfer of sulphur from thiosulphate onto a cyanide ion is catalyzes by a specific enzyme,
thiosulphatesulphide transferase:

CN
-
+ S
2
O
3
2-
SCN
-
+ SO
3
2-

cyanide thiosulphate thiocyanate sulphite

Glycine conjugation. This reaction belongs to type II conjugations, which require a prior activa-
tion of the substrate rather than of the conjugating agent. In principle, any carboxylic acid can serve as
a conjugation substrate. The mechanism of glycine conjugation may be exemplified by the formation
of hippuric acid. According to the mechanistic concept of type II conjugation reactions, the initial step
is activation of benzoic acid with the involvement of arylacyl~ScoA synthetase. Then benzoyl (or, in a
wider sense, any activated substrate in the reactions of this type) is transferred onto the glycine amino
group. This process is catalyzed by acyl-N-glycine transferase, which is specific to acylation of only
glycine, barring other amino acids.
Similarly, glycine conjugates of other compounds are formed: aromatic acids (nicotinic), phenyl-
substituted acetic acids (phenylacetic and hydratropic), steroid acids (cholic and deoxycholic).
Glutamine conjugation is a rare variety of conjugation, distinctly observable in patients with
phenylketonuria. In normal humans, the glutamine conjugation of xenobiotics has never been reported.
Factors affecting drug metabolism
Drug metabolism is affected by a variety of factors. These include genetic, age- and organ-speci-
fic, neuroendocrine, environmental factors, and the manner a drug has been administered. The rate at
which a drug supplied to the organism is metabolized is dependent on the number of enzymes invol-
ved in modification and conjugation of the drug. In enzymophathies associated with defective enzymes
that are involved in drug metabolism, a decrease in the drug metabolism rate is observed. Molecular
diseases due to a defective UDP-glucuronosyltransferase are known. These molecular diseases are
characterized by the disturbed glucuronide conjugation not only of bilirubin, but of other endogenous
substrates and drugs too. For this reason, the prescription of sulphanilamides, salicylates, and phenol-
derived preparations, which are metabolized by glucuronide conjugation, leads to aggravated symp-
toms of the disease; even normal doses of these drugs produce a negative effect.
The age is an important factor in drug metabolism. In neonates and infants, the enzymic appara-
tus of xenobiotic metabolism is poorly developed. As the young organism develops, the physiological
enzymic deficiency disappears, while hereditary enzymopathies in adult humans persist.
Liver is the major organ responsible for drug metabolism. Environmental factors such as light,
ambient temperature, radiation have been noted to influence the drug metabolism. The action of these
factors is accomplished indirectly, via the neuroendocrine system. Over 200 preparations are known to
be drug metabolism enzymes, primarily microsomal ones. They include butadion (antiinflammatory),
amidopyrine (analgesic), novocain (local anesthetic), ethanol, and others. Phenobarbital (soporific)
121
acts as the most powerful inducer. It drastically enhances the synthesis of microsomal oxidation enzy-
mes in the liver by affecting the genetic apparatus of the liver cells. Phenobarbital elicits the synthesis
of UDP-glucuronosyltransferase and facilitates the conjugation stage in the metabolism of various ma-
terials.
Inducers for drug metabolism enzymes are biogenic preparations such as thiamine, riboflavin and
their coenzymes, carnitine, pantothenic acid, androgens, and anabolic steroids; preparations of proges-
terone and estrogens inhibit these enzymes.

LECTURE 30

Subject: BIOCHEMISTRY OF CONNECTIVE TISSUE

Connective tissue is a system of insoluble protein fibers embedded in a continuous matrix called
the ground substance. It is widely distributed in the body, composing, in addition to the dermis, the
tendons, ligaments, cartilage, and matrix of bone. Its chief function is supportive and is performed by
the fibrils of insoluble proteins collagen and elastin.
The characteristics of connective tissues depend on the proportions of collagen and elastin as well
as the amount of ground substance. The Achilles tendon, for instance, a tissue of great strength, is 32%
collagen and 2.6% elastin. The ligamentum nuchae, an elastic tissue, contains 32% elastin and only
7% collagen. Although cartilage is rich in ground substance, the areolar connective tissue contains
little of the liquid matrix.
Collagen is the most abundant protein in the animal world. It serves as an extracellular frame-
work for all multicellular animals. It is most abundant in fibrous connective tissue, but also is present
in some form in practically every tissue. Examples are the ropes and straps in tendons and liga-
ments, the woven sheets in skin and fascia, porous membranes (glomeruli of the kidney), the suppor-
ting structure of bones and dentin (with embedded calcium salts, etc.), lubricating fluid with proteo-
glycans in cartilage, bone joints, and intervertebral disks, and special tissues requiring strength yet
with unusual properties, such as fatigue-resistant heart valves, and translucency, as in the cornea and
lens of the eye. At least five different types of collagen molecules have been found in the tissues of
higher animals, each apparently adapted for special extracellular functions. There are variations in
hydroxylation, glycosylation, and cross-linking. These are related to factors such as tissue types, hor-
monal factors, and even age. Abnormalities may occur in the structure and biosynthesis of collagen,
leading to a variety of human diseases.
The protein collagen, which represents almost 30% of the total body proteins, is formed by fibro-
blasts embedded in the connective tissues. The prior formation of small soluble subunits can be produ-
ced by the cell in accord with the usual processes of protein synthesis. These subunits presumably lea-
ve the cell and aggregate in a regular manner in the extracellular fluid or ground substance. The solu-
bilized collagen is then called tropocollagen and is considered to be the subunit of the mature fibers.
Tropocollagen is a highly asymmetrical molecule, a relatively rigid rod, with a molecular weight
of approximately 300,000 and dimensions of 150 to 28,000 nm. It is composed of three polypeptide
chains, each in a left-handed helical conformation having only three residues per turn. Helices of this
type have their amide and carbonyl groups extending perpendicular to the polypeptide chains so that
two of every three such groups may participate in inter-chain hydrogen bonding. The three helices to-
gether then wind is a right-handed coil (super-helix). The inter-chain hydrogen bonds appear to be the
primary linkage holding the structure together.
Collagen is unique in its high concentration of proline and glycine. These prevent the formation
of the usual -helical conformation, which requires the presence of 3.6 amino acid residues per turn.
Instead, collagen has 3 amino acid residues per turn. Each of the constituent polypeptides has 1000
residues and a molecular weight of approximately 95,000.
In connective tissues such as bone, mature dermis, tendon, and dentin, collagen has two identical
polypeptide chains called the 1-chains and a third chain of different composition called the
2
-chain.
This type of collagen is referred to as
1
(I)
2

2
. However, the composition of -chains varies in other
connective tissues. For example, in cartilage the collagen is
1
(II)
3
, meaning that the three polypepti-
des are identical but that the
1
is different from
1
(I) and also the
2
just mentioned.
The primary structure of these polypeptides consists of alternate regions of highly polar amino
acids separated by regions rich in glycine, proline, and hydroxyproline. The results of amino acid se-
122
quencing suggest the separation of polar region by nonpolar segments that average about 15 residues
in length. Summarization of information now available shows that the nonpolar regions have the follo-
wing sequence: gly-hydroxypro-ala-gly-pro-ala-gly-hydroxypro. Significant in this sequence are the
triads gly-hydroxypro-ala, gly-pro-hydroxypro, and gly-hydroxypro-pro. For the left-handed helix to
exist, every third residue must be glycine. It is now clear that the
1
- and
2
-chains are not synthesized
in their final form as found in fibroblasts. For example, neither hydroxyproline nor hydroxylysine is
incorporated directly; there are no codons for these two growing polypeptide. Hydroxylation requires a
proline or lysine hydroxylase, ascorbic acid, ferrous iron, and oxygen. The need for ascorbic acid
(vitamin C) in these reactions is evidence for the profound effect of scurvy on connective tissue forma-
tion and wound healing.
Proline + -ketoglutarate + O
2
+ Fe
2+
4-hydroxyproline + Fe
3+
+ CO
2
+ succinate

Hydroxyproline is a major component of the protein collagen. Hydroxyproline and proline play
key roles for collagen stability. They permit the sharp twisting of the collagen
helix.http://en.wikipedia.org/wiki/Hydroxyproline - cite_note-3#cite_note-3
Nascent collagen polypetyde chains are also modified by the insertion of carboxyhydrate. Thus in
the
1
-chains lysine-10
3
is hydroxylated and then glycozylated by the sequential addition of galactose
(UDP-galactose) and glucose (UDP-glucose). The side chains are peptide-hydroxylysine-galactose-
glucose. Peptide-hydroxylysine-galactose also is present.
The extracellular aggregation of tropocollagen molecules involves an orderly parallel alignment
of individual molecules to create a quarter-staggered effect. In tissue the newly formed tropocollagen
is readily extractable with neutral salt solutions because little or no cross-linking has occurred. On
aging the collagen becomes increasingly cross-linked and no longer readily extractable. Collagen is
unique in its transformation into gelatin on heating.
Elastin, is a protein having distinctly elastic properties as well as high mechanical strength. Com-
pared with that of collagen, knowledge of elastin remains meager. Elastin fibers consist of two compo-
nents: one is microfibrillar, about 1100 nm in diameter, and the other is amorphous. Elastin has a low
content of polar side chains, especially of the basic and acidic amino acids. Like collagen it has a high
content of glycine (27%) and proline (13.5%). In contrast, however, there is little hydroxyproline, no
hydroxylysine a preponderance of nonpolar amino acids, i.e. valine, alanine, leucine, and isoleucine.
Elastin is also insoluble in all solvents that do not change its chemical nature. It is not, however, con-
verted to gelatin, as is collagen.
Elastin is probably composed of long-chain precursor molecules (tropoelastin) that are polymeri-
zed by a system of cross-links. Distinctive amino acid derivatives have been obtained from elastin and
shown to have a cross-linking function. Two of these are called desmosine and isodesmosine. In the
structure of desmosine four units of lysine are required to produce the compound having a pyridine
nucleus. Three lysines are first converted to the -semialdehydes (allysine). Then the three allysines
condense with an intact peptide-bound lysine to produce the pyridine nucleus. Desmosine and an iso-
mer called isodesmosine are capable of linking together two, three, or four polypeptide chains.
Reticulin. Third fibrous protein found in connective tissues is known as reticulin, which is the
component protein of the reticular fibers. This protein resembles collagen in terms of its amino acid
composition. It appears to be distinctive, however, because of its association with lipids and carbohy-
drates.

Proteoglycans or ground substance (mucopolysaccharides)
Ground substance, a proteoglycan, may be viewed as a modified dialysate of plasma. It contains
some proteins derived from plasma by a certain degree of capillary permeability. The fluid matrix is
unique because of its high content of glycosaminoglycans or mucopolysaccharides. In fact, up to as
much as 95% carbohydrate may be present. The mucopolysaccharides present are hyaluronic acid and
chondroitin sulfates A, B, and C. These polysaccharides are synthesized by the fibroblasts.
A good example of ground substance is the lubricating fluid of all joints, called the synovial fluid.
Hyaluronic acid is an unbranched polymer of high molecular weight (I to 1.510) of a repeating
disaccharide consisting of D-glucuronic acid and N-acetyl-D-glucosamine.
The chondroitin sulfates are sulfated polysaccharides. Chondroitin sulfate A, now termed chon-
droitin-4-sulfate, is a polysaccharide of glucuronic acid and N-acetylgalactosamine bearing a sulfate
ester group on carbon-4 of amino sugar, whereas chondroitin-6-sulfate (chondroitin sulfate C) is sulfa-
123
ted on carbon-6 of the amino sugar. Chondroitin sulfate B, now called dermatan sulfate, is similar to
chondroitin sulfate A except that the uronic acid is of the sugar L-idose rather than D-glucose.
Mucopolysaccharides are believed to be complexed with protein found in the ground substance.
The chondroitin sulfates combined covalently with a single protein core (M.m.=89,000) to produce a
total unit of large molecular weight (3 to 610
6
). Therefore the chondroitin sulfates are a special type of
glycoproteins and are usually called proteoglycans. The oligosaccharide chains are attached to the sin-
gle polypeptide chain at its seryl residues. A polysaccharide made up of a repeating disaccharide in
which L-iduronic acid replaces D-glucuronic acid. These polysaccharides are found in connective tis-
sues in their sulfated forms.
Two other proteoglycans (mucopolysaccharides) are found in some tissues: keratosulfate (kera-
tin) I and II in the cornea, cartilage, spinal disks, and bone; and heparin in liver, lung, arterial walls,
mast cells. The repeating disaccharide unit in the keratosulfates is mainly D-galactose-N-acetyl-D-glu-
cosamine-6-sulfate. In heparin and heparin sulfate the repeating disaccharide unit is D-dlucuronic
acid-2-sulfate, N-acetyl-D-dlucosamine-6-sulfate, with variable amounts of N-sulfo derivatives of D-
glucosamine, rather than N-acetyl-D-glucosamine and of iduronic and glucuronic acids.
The biosynthesis of proteoglycans probably occurs by a sequential addition of monosaccharides
to the core polypeptide chain. Each monosaccharide is added to the growing polysaccharide chain by a
specific glycosyl transferase using the appropriate nucleotide-bound carbohydrate. This process may
occur in the smooth endoplasmic reticulum and the Golgi apparatus. The production of proteoglycan
probably terminates in an unsulfated glycoprotein such as the chondroitin that is found in corneal tis-
sue. The final step in the synthesis of the sulfated proteoglycans is the sulfation process itself. Sulfa-
tion requires a mixed phosphosulfate anhydride: 3-phosphoadenosine-5-phosphosulfate, or PAPS.
The proteoglycans (mucopolysaccharide-protein complexes) play numerous roles. They have the
particular ability to bind water and cations (even Na
+
and K
+
), forming a gel. Hyaluronic acid is an im-
portant constituent of vitreous humor and the umbilical cord. It imparts the lubricative quality to the
synovial fluid of the joints. Mucopolysaccharides help prevent the invasion of the body through the
skin by disease-producing microorganisms. Many bacteria produce an enzyme, hyaluronidase,that
catalyzes the depolymerization of mucopolysaccharides, thereby increasing the invasive capability of
the bacteria. Hyaluronidase is a -glucosaminidase and hydrolyzes the -1,4-glycosidic bond between
N-acetylhexosamine and the neighboring uronic acid. This enzyme is also found in spermatozoa and
may be functional in facilitating the penetration and thus the fertilization of the ovum. Highly purified
hyaluronidase is used clinically for the intradermal administration of large volumes of fluid when
intravenous injections are contraindicated.
The composition of ground substance changes with aging. In children chondroitin sulfate A
predominates, whereas in the adult chondroitin sulfate C is major component.
Cartilage is also a connective tissue and contains collagen, some noncollagenous protein, and
ground substance. Fibers of elastin are present in elastin cartilage, tending to give added flexibility to
this tissue. Vitamin D is quite possibly related to the development and health of cartilage, in addition
to being involved in the normal conversion of cartilage to bone.
Chronic rheumatouid arthritis in humans is characterized by disruption of collagenous structures.
Recent investigations have indicated that this may result from an excessive production of the enzyme
collagenase by cells of the proliferating synovium. Several investigators reported high collagenase
activity in synovial specimens from patients with rheumatoid arthritis, whereas none was demonstra-
ble in specimens from control subjects. The amount of collagenase activity was directly proportional
to the degree of local and systemic disease activity. This could account for the destruction of collagen
in and about the joints, tendons, capsules, ligaments, cartilage, and bone in patients with this type of
arthritis. A more recent hypothesis regarding the effects of arthritis on joint function suggests a depo-
lymerization of hyaluronic acid by the superoxide radical. It is proposed that superoxide is released by
phagocytic cells that have entered the synovial fluid. Superoxide dismutase derived from bovine red
blood cells is being investigated as a potential therapeutic agent.

LECTURE 31

Subject: BIOCHEMISTRY OF BONE AND TEETH

124
Bone. The organic matrix of bone or cartilage is the supporting lattice in which the bone salts are
deposited to from a rigid structure. The cartilaginous matrix is flexible and extremely strong. Collagen
is the principal protein present in bone. Collagenous fibers, formed by the osteoblasts, mature in the
mucopolysaccharide-containing ground substance. The ground substance varies in consistency from
interstitial fluid to a thick gel, and forms the communicative medium between the tissue fluid and blo-
od, thereby permitting an exchange of ions and other substances.
Analysis of bone ash reveals a preponderance of calcium, a small amount of sodium, and less
magnesium and other cations. As a matter of fact 99% of the bodys calcium is located in bone. The
anions are chiefly phosphate, with some carbonate, citrate, and small amounts of chloride and fluoride.
In general, the mineral of bone is in form of a hydroxyapatite crystal structure having the following
formula: 3Ca
3
(PO
4
)
2
Ca(OH)
2

This formula accounts for the calcium and phosphate but not for the carbonate, citrate, and small
amounts of chloride, fluoride, and other ions that are occasionally found. The belief was formerly held
that bone salt was an apatite of changing composition and that these other elements or ions were intro-
duced and withdrawn as they fluctuated in concentration in the blood plasma. Now there is evidence
that these other salts are present in an intercrystalline semiliquid medium, which allows the transport
of materials from the blood to the bone and vice versa. These two phases of the mineral structure of
bone have quite different physical, chemical, and probably physiologic properties. The hydroxyapatite
crystals are subject to rapid ion-exchange reactions at their surfaces. They are extremely-small and
thus have an enormous surface area. The intercrystalline fraction is far smaller in amount than the
crystalline fraction, about 4% as great. However, it is much more soluble, and its calcium and other
elements readily exchange with those of the blood.
The hardness and rigidity of bone are attributable chiefly to the hydroxyapatite, whereas elasticity
and toughness are attributable to the proteins.
Composition of bone:
Ash 71.0%
Water 8.2%
Collagen 18.6%
Fat 0.1%
Sugars 0.2%
Proteins 1.2%
Total: 99.3%
Citrate has been discovered as a constituent of human bone, to the extent of about 1% of the dry
weight of bone, representing as much as 70% of the bodys citrate content. The function of bone citra-
te is as yet unknown; it may play a special role in the metabolism of calcium by virtue of its power to
bind calcium. The calcium-citrate complex is soluble and diffusible but is un-ionized.
Ossification, the formation of bone salt, requires that the concentration of calcium and phosphate
must exceed the saturation point at the site of deposition.
Two enzymes are involved: glycogen phosphorylase and alkaline phosphatase. The latter hydro-
lyzes the glucose 1-phosphate formed by glycogenolysis and yields high local concentrations of inor-
ganic phosphate. It may also hydrolyze other phosphoric acid esters that may be available, e.g., glyce-
rophosphates and nucleotides. The phosphate that is found in high concentration wherever bone is for-
med is probably produced by the osteoblasts; the concentration of phosphate ions is raised locally near
these cells as a result of enzyme action. The un-ionized calcium is partly diffusible and partly not. The
nondiffusible calcium is largely that fraction combined with protein, whereas the calcium-citrate com-
plex forms most of the un-ionized diffusible part. Probably the chondroitin sulfates of growing bone
unite with calcium to provide a local surplus of available calcium. Normally the concentrations are
such that the product of ionic calcium and phosphate is about 36 to 40 mg/100 ml of plasma. Products
above 40 mg are found when bone growth or healing is taking place, whereas products below 40 mg
generally are seen in active rickets and in other conditions in which bone formation is not occurring
properly. When the local concentration of calcium and phosphate ions is increased beyond the satura-
tion point, calcium phosphate is produced. This is changed to hydroxyapatite in a series of steps.
Another enzyme, carbonic anhydrase, may be involved in the deposition of calcium salts in bo-
nes and teeth. This enzyme apparently makes available the carbonate ion for the formation of the bone
salt, carbonate hydroxyapatite. The long axes of the bone salt crystals are invariably oriented with the
long axes of the collagen fibrils and are arranged around the cross-banding of the fibrils, forming a
type of sheath.
125
Note that the bone calcium is in equilibrium with the calcium of the blood. Consequently the
blood calcium can be kept at a fairly steady concentration by a slight shift of calcium from the bones
to the blood or vice versa. The concentration of total calcium in the blood is approximately 10 mg/100
ml (2.5 mmol/L). If the level of blood calcium ions diminishes, calcium is derived from the inter-crys-
talline material. More calcium is dissolved into the surrounding fluid and enters the blood. This pro-
cess usually keeps the level at about 7 mg/100 ml. The remaining 3 mg, more or less are supplied in
just the amount needed by means of a feedback mechanism under the control of the parathyroid
glands.
Osteoporosis. In patients with osteoporosis in bone calcium salts decrease. In rickets the amount
of calcium phosphate in the bones is much below normal. This deficiency is usually the result of ina-
dequate vitamin D, which decreases the absorption and use of calcium and phosphorus. Since the con-
centration of minerals in the bones is low, the bones become less rigid and consequently bend, resul-
ting in bowlegs or other deformities. Vitamin A deficiency retards the growth of bone, particularly en-
dochondral bone formation in rats. If the deficiency is established very early in life, skeletal growth is
inhibited considerably before the effect on total increase in weight can be observed. Vitamin C also is
essential to bone development specifically the formation of tropocollagen. In skurvy there are lesions
of the epiphyseal junctions of growing bones. Subperiosteal hemorrhages are likely to occur in both
growing and adult bone. Rarefaction of the alveolar bone leads to loosening of the teeth; dentine is
resorbed, and the gums become spongy.
Teeth. The teeth resemble bone chemically to a certain extent. Over the upper surface of the tooth
is the enamel. This is the hardest substance in the body, a property of great value for the masticating
and grinding action of the teeth. Only about 5% of enamel is water. The remaining 95% consists of
inorganic material chiefly embedded in an organic matrix.
This organic matrix of enamel is composed of a protein, resembling keratin but containing no
cystine, and a mucopolysaccharide. The inorganic material is hydroxyapatite, a calcium phosphate
with the formula Ca
10
(PO
4
)
6
Ca(OH)
2
, and possibly also containing fluoroapatite Ca
5
(PO
4
)
3
F. It is an
important constituent of tooth enamel. The greater part of the tooth is dentin, which is identical to
bone from a chemical standpoint although different histologically. Dentin protein is largely collagen,
and there is chondroitin sulfate present. The inorganic basis is again an apatite, similar to the bone salt.
Administration of labeled phosphorus is followed by rapid uptake of the tracer by developing
teeth. Once the teeth are completely formed and calcified, this continuing metabolism is reduced to a
minimum. Thus the teeth are not drawn upon for calcium in time of need, as are the bones.
Vitamins A, C, and D are all necessary for proper tooth development and calcification. Lack of
vitamins A and C affects the functional activities of the formative cells. Deficiency of vitamin A
results in hypoplastic enamel imperfectly calcified. Lack of ascorbic affects the formation of the orga-
nic matrix of dentin, as it does in bone. Vitamin D not only aids in the absorption of calcium but also
apparently promotes the deposition of calcium and phosphorus in teeth.
Dental caries. When the enamel breaks and the underlying dentin exposed, dental caries deve-
lops. The cause of this formation of tooth cavities has been a matter of dispute for years and is still un-
settled. Dental caries is one of the most widespread of human diseases, and a tremendous amount of
investigation has been instituted to determine the cause and the effect a cure. In caries the enamel and
dentin are dissolved by chemical action and washed away, thus producing a cavity. Demineralization
occurs for several reasons, but the most important cause of tooth decay is the ingestion of sugars.

126


Tooth cavities are caused when acids dissolve tooth enamel:
Ca
10
(PO
4
)
6
(OH)
2
(s) + 8H
+
(aq) 10Ca
2+
(aq) + 6HPO
4
2-
(aq) + 2H
2
O(l)
The formation of cavities in the teeth is not only a source of pain and discomfort, necessitating
dental attention, but also is likely to lead to interference with mastication and thus with proper nutri-
tion. Furthermore, infections processes occurring in cavities may result in absorption of toxins or lead
to secondary infections in other parts of the body. Local bacterial factors are entirely or at least chiefly
responsible. Food particles lodged between the teeth or in recesses in the surface of teeth become bre-
eding spots for bacteria, forming bacterial plaques. If they are not removed promptly, enough organic
(e.g. lactic) acids are produced to dissolve mineral constituents from the enamel and dentin to form a
cavity and thus dental caries. Local pH values as low as 4.0 to 5.0 on the tooth surface lend support to
this view. Foods particularly rich in easily fermentable sugars (sucrose, glucose, and fructose), espe-
cially sticky sugars of candies and certain pastries, are most likely to lead to the formation of caries.
Streptococcus mutants found to be highly cariogenic, as were several species of lactobacilli and acti-
nomyces.
The influence of fluoride must be emphasized. If the amount of fluoride in drinking water is ade-
quate (1.0 to 1.2 ppm), the enamel seems to be more resistant to the development of dental caries.
Note that fluoride is probably most effective if it is present during the period of tooth development.
The fluoride content of the surface layers of enamel is normally approximately 10 times higher than
that of the layers near the dentin enamel function. It is hypothesis that fluoride acts as an enzyme inhi-
bitor, thus interrupting the chain of fermentative reactions and preventing the formation of organic
acids in proximity to the enamel.
Another view is that some component or components of saliva play a role in the prevention of
dental caries. Various proteins present in saliva have been alleged to exert anticariogenic effects.
Some propose that they act as buffers of the organic acids produced in the mouth by the fermentation
of sugars. Other, believe that the effect is immunological, certain proteins acting as antibodies. Indeed,
there are claims that a group of arginine peptides, termed sialins, are the principal agents in saliva con-
ferring resistance to caries. Another cariostatic peptide is alleged to control the incorporation of cal-
cium and phosphorus into teeth. It has been given the name statherin.
Thus vigorous biochemical and immunological research continues today in an effort to determine
the basic mechanisms involved in the prevention and control of dental caries, one of humankinds and
most prevalent chronic diseases.
Gingivitis ("inflammation of the gum tissue") is a term used to describe non-destructive
periodontal disease. The most common form of gingivitis is in response to bacterial biofilms (also
called plaque) adherent to tooth surfaces, termed plaque-induced gingivitis, and is the most common
form of periodontal disease. In the absence of treatment, gingivitis may progress to periodontitis,
which is a destructive form of periodontal disease. While in some sites or individuals, gingivitis never
progresses to periodontitis, data indicates that periodontitis is always preceded by gingivitis.
127
The etiology, or cause, of plaque-induced gingivitis is bacterial plaque, which acts to initiate the
body's host response. This, in turn, can lead to destruction of the gingival tissues, which may progress
to destruction of the periodontal attachment apparatus. The plaque accumulates in the small gaps
between teeth, in the gingival grooves and in areas known as plaque traps: locations that serve to
accumulate and maintain plaque. Examples of plaque traps include bulky and overhanging restorative
margins, claps of removable partial dentures and calculus (tartar) that forms on teeth. Although these
accumulations may be tiny, the bacteria in them produce chemicals, such as degrative enzymes, and
toxins, such as lipopolysaccharide (LPS, otherwise known as endotoxin) or lipoteichoic acid (LTA),
that promote an inflammatory response in the gum tissue. This inflammation can cause an enlargement
of the gingiva and subsequent pseudopocket formation. The primary etiology (cause) of gingivitis is
poor oral hygiene which leads to the accumulation of a mycotic and bacterial matrix at the gum line,
called dental plaque. Other contributors are poor nutrition and underlying medical issues such as
diabetes.
Microbic plaque calcifies to form calculus, which is commonly called tartar. Calculus above and
below the gum line must be removed completely by the dental hygienist or dentist to treat gingivitis
and periodontitis. Although the primary cause of both gingivitis and periodontitis is the microbic
plaque that adheres to the tooth surface, there are many other modifying factors. A very strong risk
factor is one's genetic susceptibility. Several conditions and diseases, including Down syndrome,
diabetes, and other diseases that affect one's resistance to infection also increase susceptibility to
periodontitis.
In some people, gingivitis progresses to periodontitis - with the destruction of the gingival fibers,
the gum tissues separate from the tooth and deepened sulcus, called a periodontal pocket. Subgingival
microorganisms (those that exist under the gum line) colonize the periodontal pockets and cause
further inflammation in the gum tissues and progressive bone loss.
Dental plaqueis a biofilm, usually colorless, that develops naturally on the teeth. It is formed, as
in any biofilm, by colonizing bacteria trying to attach itself to a smooth surface (of a tooth) It has been
also speculated that plaque forms part of the defense systems of the host by helping to prevent
colonization by microorganisms which may be pathogenic.
The film is soft enough to come off if scraped with a fingernail. If not removed it starts to harden
within 48 hours; in about 10 days the plaque becomes dental calculus (tartar), rock-hard and difficult
to remove.
Dental plaque can give rise to dental caries (tooth decay)the localised destruction of the tissues
of the tooth by acid produced from the bacterial degradation of fermentable sugarsand periodontal
problems such as gingivitis and chronic periodontitis.
The mechanisms of plaque formation include:
- Absorption of proteins and bacteria to form a film on the tooth surface.
- The effect of van der Waals and electrostatic forces between microbial surfaces and the film to
create reversible adhesion to the teeth.
- Irreversible adhesion due to intermolecular interactions between cell surfaces and the pellicle.
- Secondary colonisers attach to primary colonisers by intermolecular interaction.
- The cells divide and generate a biofilm.
Components of plaque. Plaque consists of microorganisms and extracellular matrix. The
microorganisms that form the biofilm are mainly Streptococcus mutans and anaerobes, with the
composition varying by location in the mouth. Examples of such anaerobes include fusobacterium and
actinobacteria. The extracellular matrix contains proteins, long chain polysaccharides and lipids. The
microorganisms present in dental plaque are all naturally present in the oral cavity, and are normally
harmless. However, failure to remove plaque by regular tooth brushing means that they are allowed to
build up in a thick layer. Those microorganisms nearest the tooth surface convert to anaerobic
respiration; it is in this state that they start to produce acids.
- Acids released from dental plaque lead to demineralization of the adjacent tooth surface, and
consequently to dental caries. Saliva is also unable to penetrate the build-up of plaque and thus
cannot act to neutralize the acid produced by the bacteria and remineralize the tooth surface.
- They also cause irritation of the gums around the teeth that could lead to gingivitis,
periodontal disease and tooth loss.
- Plaque build up can also become mineralized and form calculus (tartar).
128
Dental fluorosis is a health condition caused by a child receiving too much fluoride during tooth
development. The critical period of exposure is between 1 and 4 years old; children over age 8 are not
at risk. In its mild form, which is the most common, fluorosis appears as tiny white streaks or specks
that are often unnoticeable. In its severest form, which is also called mottling of dental enamel it is
characterized by black and brown stains, as well as cracking and pitting of the teeth. The severity of
dental fluorosis depends on the amount of fluoride exposure, the age of the child, individual response,
as well as other factors including nutrition. Although water fluoridation can cause fluorosis, most of
this is mild and not usually of aesthetic concern. Severe cases can be caused by exposure to water that
is naturally fluoridated to levels well above the recommended levels, or by exposure to other fluoride
sources such as brick tea or pollution from high fluoride coal. Dental fluorosis occurs because of the
excessive intake of fluoride, either through fluoride in the water supply, naturally occurring or added
to it; or through other sources. The damage in tooth development occurs between the ages of 3 months
to 8 years, from the overexposure to fluoride. Teeth are generally composed of hydroxyapatite and
carbonated hydroxyapatite; when fluoride is present, some fluorapatite is generated. Excessive
fluoride can cause white spots, and in severe cases, brown stains or pitting or mottling of enamel.
Fluorosis cannot occur once the tooth has erupted into the oral cavity. At this point, fluorapatite is
beneficial because it is more resistant to dissolution by acids (demineralization). Although it is usually
the permanent teeth which are affected, occasionally the primary teeth may be involved. The
differential diagnosis for this condition may include Turner's hypoplasia (although this is usually more
localized), some mild forms of amelogenesis imperfecta, and other environmental enamel defects of
diffuse and demarcated opacities.
LECTURE 32

Subject: BIOCHEMISTRY OF SALIVA

Saliva is the mixed secretion of the parotid, submaxillary, sublingual, and buccal glands. It con-
tains 99.3% to 99.7% water and has a specific gravity of 1.002 to 1.008. Approximately 1500 ml is be-
lieved to be the daily secretion in man. The secretion of saliva is entirely under the control of the ner-
vous system. A variety of stimuli cause an increased flow by reflex stimulation. This is true whether
the stimulus is psychic (sight, smell, or thought of food), mechanical (chewing), or chemical (action of
acids, salts, etc. on the taste buds). There seems to be no hormonal control of salivary secretion.

1 Parotid gland; 2 Submandibular gland; 3 Sublingual gland

Saliva is almost colorless and rather viscid, and if a quantity of saliva is a vessel is exposed to air,
the surface becomes covered with an incrustation consisting of calcium carbonate with a small propor-
tion of organic matter. The reaction of the saliva of a given individual is not constant. Resting saliva is
slightly acidic, pH 6.4 to 6.9, whereas saliva obtained during active stimulation of the glands is neutral
to slightly alkaline, pH 7.0 to 7.3.
The solid constituents of saliva comprise albumins, globulins, mucins, enzymes, urea, uric acid,
glucose, and inorganic salts. The salivary mucins are glycoproteins and yield 30% to 45% carbohydra-
te. Ovine submaxillary mucin, for example, contains approximately 45% carbohydrate and has about
800 disaccharide units per protein molecule. The disaccharide units are known to be N-acetyl-
neuraminyl (26) N-acetylgalactosamine. The disaccharide units are attached to every sixth amino
129
acid residue of the protein component a single polypeptide chain. The protein portion is rich in
threonine, with a low isoelectric point, about 3.5, probably because of the carbohydrate moiety. This
material has a relatively high viscosity and a high degree of hydration, which in part account for the
protective and lubricating functions described below. The inorganic components differ greatly in con-
centration from those of blood serum, but the nonprotein nitrogen-containing constituents (urea, uric
acid, NH
4
+
salts) appear to bear some relation to these same constituents in the blood. Amino acids and
glucose occur in extremely small amounts in the saliva of healthy individuals (11 to 30 mg glucose per
100 ml). Both salivary cholesterol and lipid phosphorus values are very low as compared with blood.
The salivary glands therefore appear to be quite selective in secretary action.
The chief inorganic ions present are K
+
, PO
4
-
, and Cl
-
, with smaller amounts of Na
+
, Ca
2+
, and
SO
4
-
. Some of these may combine to form insoluble precipitates. This may be added by changes in the
pH brought about by decomposing food material left between the teeth or by the loss of carbon dioxi-
de, held in solution in the saliva, as soon as it meets atmospheric conditions. Thus tartar may be for-
med. This consists chiefly of calcium carbonate and phosphate. Salivary calculi sometimes are formed
in the ducts and the similar in composition to tartar (namely, Ca
3
[PO
4
]
2
or CaCO
3
). It is usually stated
that a clump of bacteria or a foreign body establishes a nucleus around which the precipitation of these
salts occurs. However, calcium oxalate may be the precipitated salt, which, together with mucin and
globulin, may form the calculus. Increased acidity is necessary for oxalate calculus formation.
Functions of saliva. Saliva has a digestive function because of the enzymes present, but it also
has other functions. It moistens and lubricates the food, permitting it to be swallowed easily. Saliva
holds the taste-producing substances in solution and so brings them into contact with the taste buds. It
dilutes salts, acids, etc., thereby protecting the mucosa and, to some extent the teeth. It also has a clea-
nsing action on the teeth, gums, and buccal mucosa. It owes its viscous and lubricating property to its
content of mucin. This protein is present as an alkaline salt, which is soluble at the pH of saliva but is
precipitated on acidification. It is one of the chief buffers present in saliva. A major function of epithe-
lial mucins in general is the protection of the mucosal lining of the mouth, the gastrointestinal tract,
and the inner surfaces of other body cavities. They form water-soluble films. In the stomach the acidi-
ty probably results in the formation of insoluble gels and although the mucins are not completely resis-
tant to proteolytic enzymes, the action is slow and thus there is considerable protection. Some authori-
ties maintain that saliva has an excretory function, since certain elements and drugs are found in it af-
ter administration. Among these are mercury, lead, and potassium iodide. Any part of them lost in ex-
pectoration could be considered excreted, but some of the part swallowed may be reabsorbed. Hence it
is difficult to see how these elements can be called a true excretion. The same is true of the traces of
urea, uric acid, and ammonium salts ordinarily found in saliva.
The parotid and submaxillary salivary glands have been implicated in the deiodination of the hor-
mone thyroxine and hence in the regulation of the thyroxine level of the blood. The iodine thus relea-
sed is excreted in the saliva and is reabsorbed in the small intestine for reuse, completing an iodine
cycle.
Enzymes. The principal enzyme of human saliva is an o-amylase (ptyalin). There are also traces
of possibly a maltase, catalase, urease, protease, and others.
Some investigators maintain an important function of salivary o-amylase is a cleansing agent for
the oral cavity; i.e. digests starch particles or pastes left in or near the teeth. Salivary amylase is 1,4-
glycosidase and serves to hydrolyze only 1,4-glycosidic bonds found in starch and glycogen. The spe-
cificity of this enzyme is such that it can only hydrolyze alternate glucosidic bonds. Consequently the
result of the amylolytic process beginning in the saliva involves the conversion of starch and glycogen
into maltose and dextrin. The latter is a mixture of large-chain, branched oligosaccharides or partial di-
gestion products.

Indices of saliva
Daily volume (V) 500-1500 ml (500-1500 ml/24 h)
pH 6.07-7.9
Density () 1.08-1.32
Nitrogen-containing organic substances
Proteins 1.4-6.4 g/L
Mucine 0.8-6.0 g/L
Ammonia 0.01-0.12 g/L
Urea 0.14-0.75 g/L
130
Uric acid 0.005-0.029 g/L
Creatinine 0.005-0.750 g/L
Choline 0.005-0.036 g/L
Nitrogen-noncontaning organic substances
Glucose 0.10-0.30 g/L
Citric acid < 0.020 g/L
Lactic acid 0.01-0.05 g/L
Cholesterol 0.025-0.500 g/L
I norganic substances
Sodium (Na) 5.2-24.4 mmol/L
Potassium (K) 14-41 mmol/L
Calcium (Ca) 2.3-5.5 mmol/L
Chlorides (Cl
-
) 15.1-31.6 mmol/L
Phosphate inorganic 0.080-0.217 g/L
Bicarbonate (HCO
3
-
) 2.13-13.00 mmol/L
Fluor (F
-
) 0.8-2.5 g/L
Brom (Br
-
) 0.2-7.1 mg/L
Tiocianate (SCN
-
) 0.12-0.33 g/L
Magnezium (Mg
2+
) 0.16-1.06 mmol/l
Sulphates 0.04-0.2 g/L
Copper (Cu) 0.5-7.6 mg/L
Zinc (Zn) 0.06-0.80 g/L
Vitamins
Thiamine (B
1
) 0.7 g/dL
Riboflavin (B
2
) 5.0 g/dL
Pyridoxine (B
6
) 60 g/dL
Nicotinic acid (PP) 3.0 g/dL
Pantotenic acid 8.0 g/L
Ascorbic acid (C) 0.58-3.78 mg/L
Biotin (H) 0.08 g/dL
Phyllochinon (K) 1.5 g/dL

LECTURE 33

Subject: BIOCHEMISTRY OF MUSCLE TISSUE

Muscles form a large proportion of the active tissue of the body. In normal adults it is fully two
fifths of the body weight, but about half the metabolic, or chemical and physical, activity of the body
takes place in muscles even during rest. When the muscles are contracting, while doing work, fully
three fourths of the total metabolism can be assigned to them.
The three types of muscle-striated (voluntary) skeletal, cardiac, and nonstriated (involuntary)
smooth-differ somewhat in their chemistry, but they have the same general characteristics. In skeletal
muscle are found the following:
- water,
- proteins (albumins, globulins, nucleoproteins, myoglobin),
- lipids (cholesterol, phospholipids, triglycerides),
- extractives (soluble in hot water),
- nonnitrogenous (creatine, creatine phosphate, creatinine, inosinic acid, adenylic acid, adenosi-
ne triphosphate, glutathione, purines, pyrimidines, carnosine, anserine, choline, acetylcholine),
- enzymes,
- hormones,
- vitamins,
- inorganic salts.
Adult muscle has 72% to 78% water. As in the case of nerve tissue, the water content of the
muscle of the young and of the fetus is even higher. The solids of muscular tissue are largely protein in
131
nature, whereas those of nerve tissue are largely lipid. The total lipid of muscle amounts to only about
3% and the glycogen less than 1%, but the protein content is about 20%.
Structure of skeletal muscle. A typical skeletal muscle consists of many muscle fibers, or fasci-
cules, the structural unit of muscle. Numerous fibers are bound together by areolar connective tissue
and are arranged parallel to the line of its attachments via tendons to the bones or other structure who-
se movements they control. Typically, muscle fibers are approximately 0.01 to 0.1 mm in diameter
and from 1 to 40 mm in length. Groups of fibers are encased in a thin but tough elastic sheath, known
as the sarcolemma, beneath which are embedded numerous nuclei and mitochondria. The sarcolemma
is an electrically excitable membrane, important in the transport of motor nerve impulses. The sarco-
lemma also contains numerous longitudinal and transverse sarcotubules (transport ducts) and the sar-
coplasmic reticulum. Bundles of muscle fibers are surrounded by a semi-fluid substance, the muscle
plasma or sarcoplasm. The sarcoplasm contains the red protein myoglobin (muscle hemoglobin),
which transports oxygen from adjacent blood capillaries to the sites of oxidation in mitochondria.
Myoglobin, like erythrocyte hemoglobin can be converted to metmyoglobin by various oxidants (e.g.,
certain drugs) and reconverted to myoglobin by mechanisms similar to those present in red blood cells
(antioxidant defense systems).
Muscular fibers are made up of numerous smaller structures called myofibrils. These contain tran-
sverse alternating light (isotopic) I bands bisected by a dark Z line (membrane) and dark (anisotropic)
A bands, having a lighter central H zone. These bands are aligned in adjacent myofibrils to appear as
continuous bands, or striations, across an entire muscle fiber.
Myofibrils also contain numerous fine longitudinal lines, sometimes termed myofilaments, con-
sisting of actin (thin) filaments and myosin (thick) filaments containing a number of cross-bridges. A
single sacromere, the functional unit of the myofibril is bounded by two Z lines.
Proteins of muscle. As started before, about 20% of muscle tissue is protein. Over half the mus-
cle proteins are the structural proteins: actin, myosin, tropomyosin, and troponin. Other proteins inclu-
de myoglobin, a number of enzymes involved in the metabolism of muscle tissue, and collagen and
other proteins of connective tissue.
Myosin, a globulin, is the most abundant muscle protein. The myosin molecule consists of two glo-
bular heads, each joined to a tail-like appendage and twisted into a double-stranded o-helix. The mo-
lecular weight is approximately 500,000. The two identical major chains have molecular weights of
about 20,000 each. Several hundred myosin molecules bind together tail-to-tail to form the thick filament
of the sarcomere A band. Their double heads project at regular intervals forming the cross-bridges of the
sarcomere. The tails meet at the central H band of the sarcomere. The head portion of myosin has a
marked ability to bind with actin to form actomyosin, which has a pronounced ATPase activity.
Actin also is a globulin, with a molecular weight of about 60,000. It is a major constituent of the
thin filaments of the sarcomere. A lower molecular weight form of actin, called G-actin (M.m.=
42,000), with a globular configuration, is obtained by extracting muscle tissue with a low-ionic stren-
gth medium. If the ionic strength is increased and Mg
2+
ions and ATP are present, G-actin polymera-
zes into F-actin, a fibrous form. It has a double-helical structure resembling a double chain of beads.
F-actin combines with myosin to form actomyosin, which has the remarkable property of contrac-
tion in the presence of ATP, K
+
, and Mg
2+
, as shown in the classic experiments of Szent-Gyorgyi. He
correctly concluded that the force of muscular contraction comes from an interaction of actin, myosin,
and ATP.
Two other proteins, tropomyosin and troponin are now known to be involved in the contraction of
muscle. Tropomyosin is a double-stranded o-helical rod with a molecular weight of 70,000. In the thin
actin filaments it is intertwined between the two helical strands of F-actin.
Troponin, which is positioned at regular intervals along actin filaments, is a spherical molecule
containing three different types of subunits, with a molecular weight of approximately 70,000. It is the
third component of the thin filament. The three subunits are named according to their functions: Tn-T
binds to tropomyosin; Tn-I is the inhibiting unit; and Tn-C is the calcium-binding unit. The Tn-T subu-
nit has a molecular weight of 37,000 and contains 259 amino acid residues. Its action appears to be to
prevention of the interaction of the head of myosin molecules with actin, thus preventing the binding
to actin and ATPase activity. Contraction of the myofibril is thus prevented. Troponin-C, which binds
calcium ions, has a molecular weight of about 18,000 and contains 159 residues, a relatively large
number being aspartic and glutamic acids. Tn-C also competitively binds Mg
2+
ions.
Sequence of events during muscle contraction. Contraction of the myofibrils, indeed the sarco-
meres of the myofilaments, is initiated by nerve impulses transmitted via motor nerves across the neu-
132
romuscular junction to the sarcolemma of muscle fibers. A wave of depolarization spreads from this
point along the entire fiber and then inward through openings in the transverse tubules into sarcoplas-
mic reticulum of the myofibrils. This stimulates a release of Ca
2+
ions by some as yet uncertain mecha-
nism. The Ca
2+
ions bind to troponin-C (of the thin actin filaments), producing a change in its confor-
mation that is followed by a shift of tropomyosin, permitting the attachment of the cross-bridges of the
myosin (thick) myofilaments to the actin (thin) myofilaments. Simultaneously, energy is released by
the hydrolysis of the ATP by the ATPase action of actomyosin. This energy is used to cause a ratchet-
like movement of the myosin cross-bridges attached to the actin filaments, causing the latter to slide
along the stationary myosin (thick) filaments towards the center of the sarcomere. These results in the
shortening of the sarcomere and in turn the myofibril, the muscle fiber, and the muscle as a whole. The
foregoing events occur quite rapidly, the contraction process requiring only a 10 to 20 msec.
When the neurostimulatory phase ends, Ca
2+
ions are drawn back into the sarcoplasmicc reticu-
lum network, the cross-bridges of myosin retract, and the sarcomeres and the muscle fibers relax, retu-
rning to the resting state. The sequence may be repeated for as long as the supply of ATP and other
components essential for muscle contraction permit.
Source of energy for muscle contraction. ATP is the immediate source of energy for muscle con-
traction. However, the amount of ATP in muscle is relatively small, so a back-up reserve supply of
readily available energy is necessary. This is supplied in the form of phosphocreatine (phosphagen),
the high-energy phosphate of which is transferred to ADP by the enzyme creatine phosphotransferase
(creatine kinase or CPK) the Lohmann reaction. Mammalian skeletal muscle contains about five
times as much phosphocreatine as ATP. This still is a fairly limited supply of ATP, so replenishment
from other sources is soon necessary. Muscle glycogen is available to a limited extent, forming ATP
by means of glycolysis and the citric acid cycle. Any lactic acid formed in muscle contraction under
anaerobic conditions can be converted to glucose-glycogen in the liver and then into ATP. Free fatty
acids and ketone bodies also can be used as a source of ATP in most skeletal muscles. Gluconeogene-
sis is another source. ATP also may be derived from ADP by the muscle myokinase reaction:

2ADP ATP + AMP

The reserve supply of phosphocreatine in muscle is regenerated by the transfer of the high-energy
phosphate of ATP to creatine via the reversible action of creatine phosphokinase. The resynthesis of
ATP and phosphocreatine can be blocked by agents, such as iodoacetate, that inhibit glycolysis. Gly-
colysis thus is the ultimate major source of energy for the contraction of skeletal muscle. Other types
of muscle (aerobic), however, can use additional types of fuel efficiently, such as free fatty acids, lac-
tate, and ketone bodies.
Metabolism in other types of muscle
Cardiac muscle, which is characterized by its capability for long, sustained contractions, can rea-
dily use fuels requiring aerobic conditions, such as free fatty acids, lactate, and ketone bodies. Indeed,
there is some evidence that free fatty acids are a preferred source of energy for the myocardium. The
mechanisms of contraction of cardiac muscle are essentially the same as those described for skeletal
muscle.
Smooth muscle (involuntary) differs sharply from skeletal muscle in its structure, location in the
body, innervations, mode of action, and general functions. However, the chemical processes involved
in the contraction of smooth muscle are similar to those found in skeletal and cardiac muscle, e.g., the
requirement for calcium ions for initiation and ATP for the force of contraction.
Muscle lipids. Besides variable amounts of fat, muscle is found to contain small amounts of cho-
lesterol and larger quantities of phospholipids. Here there are definite differences among the three
types of muscle. Smooth muscle has the greatest amount of cholesterol, cardiac muscle next, and stria-
ted muscle the least. The ratio of phospholipids to cholesterol is high for skeletal and cardiac muscle
and low for smooth muscle. These findings indicate that cholesterol has some relation to the sponta-
neous muscular activity of cardiac and smooth muscle, and phospholipids are involved in some way
with the greater energy production of cardiac and striated muscle.
Extractives. If muscle tissue is ground and repeatedly extracted with hot water, a light tan fluid
with droplets of fat floating on its surface and particles of coagulated protein suspended is obtained.
When this is filtered and concentrated, a dark brown sticky material, commonly known as beef extract,
is left. This is composed of all the soluble inorganic salts and all the extractives mentioned earlier.
Aside from the small amounts of carbohydrate, amino acids, and peptides present, there is little food
value in beef extract.
133
Some of the individual extractives deserve mention at this point. One of them is inositol,
C
6
H
12
O
6
, or better, C
6
H
6
(OH)
6
. There are a number of natural isomers of inositol. The most important
one was renamed myoinositol, instead of mesoinositol.
At least four peptides have been isolated from muscle extracts. Carnosine (|-alanylhistidine), an-
serine (|-alanyl-1-methylhistidine), and |-alanyl-3-methylhistidine are compounds found only in ske-
letal muscle of vertebrates. The fourth peptide present in muscle extractives is glutathione, a tripepti-
de, with the composition glutamylcysteinylglycine. This compound is a hydrogen acceptor and as such
plays a role in tissue reactions, such as the reduction of methemoglobin. It is found in many tissues
other than muscle, especially liver, red blood cells, brain, and kidney. It is also present in the lens of
the eye and is reduced in amount when cataract occurs. Carnitine, a betaine, is also a muscle extractive
and is also found in most tissues. Its formula is as follows:

CH
3
OH O
-

| | |
CH
3
-N
+
-CH
2
-CH-CH
2
-C=O
|
CH
3

Carnitine plays a role in the oxidation of fatty acids in muscle tissue, as well as in other tissues,
by facilitating their transfer to fatty acid oxidation sites.
Other constituents. Other constituents include glycogen (0.5% to 1.0%), important as a source of
energy for the formation of ATP, traces of free amino acids, a number of enzymes, and inorganic ions,
including potassium, magnesium, and sodium as the principal cations in descending amount, and phos-
phate, sulfate, and bicarbonate as the main anions similarly.

LECTURE 34

Subject: PRODUCTION OF ENERGY IN PHOTOSYNTHETIC ORGANISMS

Photosynthetic organisms (photoautotrophs) convert luminous energy to chemical energy to be
used in the synthesis of carbohydrates and other materials. Photosynthetic cells possess an apparatus
for acquisition and conversion of energy and enzymic systems for CO
2
assimilation. Photosynthetic
organisms are diversified. They include multicellular organisms (higher green plants and their lower
forms: green, brown, and red algae) and unicellular organisms (euglenaceae, diatoms, and dinoflagel-
lata). A large group of photosynthetic organisms is represented by procaryotes (green-blue algae, gre-
en and purple bacteria). About half the overall work in the photosynthesis on the Earth accomplished
by higher green plants, and the other half, chiefly by unicellular algae.
Photosynthetic structures. In bacteria, the photosynthetic structures are represented by invagina-
ted portions of the cell membrane, which form lamellar organelles called mesosomes. Isolated mesoso-
mes produced from disintegrated bacteria are called chromatophores; they accommodate the light-sen-
sitive apparatus of bacteria. In eukaryotes, the photosynthesis apparatus is located in special intracellu-
lar organelles, chloroplasts. Chloroplasts, similar to mitochondria, contain also DNA, RNA, and the
apparatus for protein synthesis, i.e. potentially they are capable of self-reproduction. In size, chloro-
plasts are several times as large as mitochondria. The number of chloroplasts per cell varies over a ran-
ge from 1 (in algae) to 40 (in higher plants). The green plant cells contain, alongside of chloroplasts,
mitochondria which are employed for energy generation at night time by the respiration mechanism,
like in heterotrophic cells.
Chloroplasts are of spherical or oblate (lense-like) shape. A chloroplast possesses two-outer and
inner-membranes. Stacks of flattened vesicle-like disks extend from the inner membrane. These stacks
are called the grana. The number of grana per chloroplast in algae does not exceed 1, while in higher
plants, 50 grana link with each other through membraneous bridges. The aqueous medium of the chlo-
roplast in which the grana are immersed is called the stroma. The vesicle-like structures, which the
grana are composed of, are referred to thylakoids. One granum contains 10 to 20 thylakoids.
The elementary structural and functional photosynthetic unit of the thylakoid membrane, which
contains necessary light-trapping pigments and components of the energy conversion apparatus, is
called the quantasome. This particle has a molecular mass of about 210
6
daltons and measures about
17.5 nm.
134



The simplified internal structure of a chloroplast

Stages of photosynthesis. Photosynthesis is the conversion of luminous energy to chemical ener-
gy, with the sequent use of the latter in the synthesis of carbohydrates from carbon dioxide. The ove-
rall reaction for the photosynthesis is:

6CO
2
+ 12H
2
O nhv C
6
H
12
O
6
+ 6H
2
O + 6O
2
+ 2861 kJ/ml

This process is endergonic and requires much energy. Therefore, the overall process of photosyn-
thesis is made up of two stages, commonly referred to as the light (or energetic) reaction and the dark
(or metabolic) reaction. In chloroplasts, these two stages are spatially separated: the light reaction is
carried out in the quantasomes of thylakoid membranes, while the dark reaction is accomplished ex-
trathylakoidally, in the stroma. The relationship between the light and dark reactions may be presented
schematically as:
12 H
2
O 6O
2
12 NADPH
+
+H
+
+ 6H
2
O
light ` ` 6CO
2
C
6
H
12
O
6

Light reaction 18 ATP | Dark reaction

The light reaction proceeds in the light. At this stage, the sunlight energy is converted to the ATP
chemical energy, and the energy-deficient water electrons are transferred to become the energy-rich
NADPH
2
electrons. The oxygen liberated during the light reaction is a byproduct of this process. The
energy-rich light-reaction products, ATP and NADPH
2
, are used at the sequent stage which can pro-
ceed in the dark. The reductive synthesis of glucose from CO
2
occurs during the dark reaction. The
dark reaction cannot be accomplished independently of the light reaction.
Mechanism of the light (photochemical) reaction in photosynthesis. In the thylakoidal membra-
ne, there are two photochemical centers, or photosystems, commonly denoted as photosystems I and II.
Either of the two photosystems cannot replace the other one, since functionally these photosystems are
different. The photosystems contain various pigments: green pigments, chlorophylls a and b; yellow
pigments, carotinoids; and red or blue pigments, phycobilins. In this variety of pigments, only chlo-
rophyll a is photochemically active. The other pigments play a minor role and act merely as photon
collectors (a kind of focusing lenses) or light guides to the photochemical center. The function of the
photochemical center is assigned to special forms of chlorophylls a which are: in the photosystem I,
the pigment 700 (P
700
) absorbing light at a wave-lenght of about 700 nm, and in the photosystem II, the
pigment 680 (P
680
) absorbing light at a wavelenght of 680 nm. In the photosystems I and II, there
occurs only one molecule of the photochemically active pigment, chlorophyll a, per 300 to 400 mole-
cules of light-collecting pigments. The absorption of protons by photosystem I induces a transition of
the P
700
pigment from the ground state into an excited state, P
*
700
, in which the pigment molecule
easily loses an electron. The electron loss leads to the formation of an electron hole P
*
700
according to
the scheme:
P
700
hv P
*
700
P
+
700
+ e
The electron hole is apt to easily recapture an electron. Thus, absorption of photons by the photo-
system I leads to a separation of charges: the positive charge formed as an electron hole (P
+
700
), and
135
the negatively charged electron, which is initially accepted by a special iron-sulphur protein (FeS cen-
tre), further to be either transported via a carrier chain back to P
700
(to fill the electron hole), or trans-
ferred via another carrier chain through ferredoxin and flavoprotein to the permanent acceptor
NADPH
2
. In the former instance, a closed cyclic electron transport, and in the latter instance, a non-
cyclic electron transport occurs. The return of excited electrons onto P
700
is associated with a release
of energy (transition from a higher onto a lower energy level), which is accumulated in the ATP phos-
phate bonds. This process is called photophosphorylation. Under cyclic electron transport conditions,
the cyclic photophosphorylation, and under noncyclic electron transport, respectively, the noncyclic
photophosphorylation occurs. In the thylacoids, both processes are operative, the latter one being
somewhat more complicated, since it is coupled to the operation of photosystem II.
Absorption of photons (H
+
) by the photosystem II leads to decomposition (photooxidation) of
water in the photochemical centre P
680
according to the scheme:

H
2
O hv 2H
+
+ 2e +
1
/
2
O
2
(Hill reaction)

Photolysis of water is referred to as the Hill reaction. The electrons released by water decomposi-
tion are initially accepted by the substance denoted Q (occasionally, it is also called cytochrome C
550

by its light absorption maximum, although this compound is not a cytochrome). Then from the subs-
tance Q, the electrons are transferred towards P
+
700
to fill the electron hole a carrier chain compositio-
nally similar to the mitochondrial chain.
Consequently, the lost P
700
electrons are replenished with the electrons derived from water photo-
decomposed in the photosystem II. The observed noncyclic electron flow from H
2
O to NADPH
2
,
which is produced by the interaction of the two photosystems and their coupling electron-transport
chains, formally is reverse to the respective redox potentials: E
o
=+0.81v for
1
/
2
O
2
/H
2
O and E
o
=-0.32v
for NADP
+
/NADPH
+
. In other words, the luminous energy turns the electron flow upstream. To be
noted, on the II-to-I photosystem electron transfer, a portion of electron energy is stored on the thyla-
coid membrane as a proton potential which is further converted to the ATP energy.
The mechanism by which the proton potential is produced in the electron transfer chain to be used
for the ATP formation in chloroplasts is similar to that in mitochondria. However, the photophospho-
rylation mechanism exhibits certain specific features. In a sense, thylacoids may be likened to mito-
chondria turned inside out; therefore, the direction of electron and proton transport across the thyla-
koid membrane is reversed to that observed for the mitochondrial membrane. The electrons are trans-
ferred towards the outer side, while the protons are stored within the thylakoid matrix. The matrix ac-
quires a positive charge, and the outer thylakoid membrane, a negative charge, i.e. the direction of the
proton potential is opposite to that of the mitochondrial proton potential. Another specific feature is a
substantially larger contribution of pH to the proton potential as compared with the mitochondria. The
thylakoid matrix is more acidic.
The H
+
-ATP-synthetase, denoted in chloroplasts as the complex CF
1
+ F
o
, is also oriented in
the opposite direction. Its knob (coupling factor F
1
) faces outwards, towards the chloroplastic stroma.
The protons are forced outwards from the matrix through the CF
1
+F
o
complex, and ATP in the active
centre is produced through the use of proton potential energy.
As distinct from the mitochondrial chain, there appear to be only two coupling sites in the thyla-
koidal chain; therefore three instead of two protons are required for the synthesis of one ATP molecu-
le, i.e. the molar H
+
/ATP ratio is 3:1.
Mechanism for the dark reaction in photosynthesis. The products of light reaction, ATP and
NADPH
2
, which are formed in the chloroplast stroma, are immediately used in the CO
2
synthesis of
glucose. Assimilation of carbon dioxide (photochemical carboxylation) is a cyclic process commonly
referred to as the photosynthetic pentose-phospate cycle, or Calvin cycle. Three major phases are
distinguished in the cycle:
1. fixation of CO
2
with ribulose biphosphate,
2. production of triose phosphates by reduction of 3-phosphoglycerate,
3. regeneration of ribulose biphosphate.
The fixation of CO
2
with ribulose biphosphate is catalyzed by the enzyme ribulose-biphosphate
carboxylase:
Ribulose biphosphate + CO
2
3-phosphoglycerate

3-phosphoglycerate is further reduced by NADPH
2
and ATP to glyceraldehyde 3-phosphate. This
reaction is catalyzed by the enzyme glyceraldehyde 3-phosphate dehydrogenase. Glyceraldehyde 3-
136
phosphate is readily isomerizable to dihydroxyacetone phosphate. Both triose phosphates are used in
the synthesis of fructose biphosphate (reverse reaction, catalyzed by fructose-biphosphate aldolase).
The fructose biphosphate produced is partly involved, alongside triose phosphates, in the regeneration
of ribulose biphosphate, and partly is stored as carbohydrate reserve in the photosynthetic cells.
As has been estimated, the synthesis of one glucose molecule from CO
2
in the Calvin cycle requi-
res 12 NADPH
+
+H
+
and 18 ATP (12 ATP molecules are spent on the reduction of 3-phosphoglycera-
te, and 6 molecules are used in the ribulose bisphosphate regeneration reactions). The minimal ATP-
to-NADPH
2
ratio is 3:2.
One may perceive a generality of the principles that constitute the basis for photosynthetic and oxi-
dative phosphorylations, the photophosphorylation being a kind of reversed oxidative phosphorylation:
SH
2
AH
2
ATP
1
/
2
O
2
H
2
O oxidative phosphorylation
SH
2
AH
2
H
2
O
1
/
2
O
2
Sunlight
ATP photosynthetic phosphorylation

The sunlight energy is a driving force for phosphorylation and synthesis of organic materials
(SH
2
) under photosynthesis and, vice versa, the energy of organic material oxidation is a driving force
under oxidative phosphorylation. Precisely for this reason, plants ensure existence for animals and
other heterotrophic organisms:
H
2
O ATP | H
2
O
Sunlight + AH
2
SH
2
| SH
2
AH
2
|
1
/
2

O
2
| ATP
1
/
2
O
2

Plants Animals

Carbohydrates produced by photosynthesis serve for building the carbon frameworks of numero-
us organic materials in plants. Ultimately, photosynthesis provides for the buildup of not only essential
proteins, nucleic acids, carbohydrates, lipids, and cofactors, but also numerous products of secondary
synthesis, which exhibit valuable medicinal properties (alkaloids, flavanoids, polyphenols, terpenes,
steroids, organic acids, etc.).
Photosynthesis and environment. Photosynthesis is feasible only if light, water, and carbon dio-
xide are available. In cultivated plants, the photosynthesis efficiency does not exceed 20%; usually, it
is not higher than 6-7%. The atmospheric occurrence of CO
2
is about 0.03% (by volume); an increased
CO
2
content (to 0.1%) is favourable for photosynthesis and plant productivity. It is therefore expedient
to use hydrogen carbonates as fertilizer additives in plant cultivation. However, an atmospheric CO
2

content higher than 1.0% exerts a deleterious effect on photosynthesis. Land plants assimilate 3% of
the Earths atmospheric CO
2
, i.e. about 20 thousand million tons per year. The byproduct of photosyn-
thesis, oxygen, is of vital importance for higher organisms and aerobic microorganisms. In preserving
the plant kingdom we are saving life on Earth.




















137
LITERATURE

1. Bhagavan N.V. Medical Biochemistry. Fourth Edition, Academic Press, 2002.
2. Campbell P.N., Smith A.D. Biochemistry illustrated. International Edition, 2000.
3. Cohn R.M., Roth K.S. Metabolic diseases. A Guide for Early Recognition. W.B.Saunders
Company, Philadelphia, London, Toronto, Tokyo, 1983.
4. Dati Francesco, Metzman Erwin. Proteins. Laboratory testing and clinical use. Includes
index. Holzheim, Germany, 2005.
5. Dawson Rex M.C., Elliott Daphne C., Elliott William H., Jones Kenneth M. Data for
Biochemical Research. Third Edition. Clarendon Press, Oxford, 1986.
6. Gavriliuc Ludmila A. Biochemistry: lectures for students of Medical Departments. Chisinau,
2001.
7. Emery A.E.H., Mueller R.F. Elements of Medical Genetics. ELBS Longman Group UK Ltd.,
1992.
8. Lehninger A.L., Principles of Biochemistry, The Johns Hopkins University School of
Medicine, Worth Publishers Inc., 2007.
9. Marshall William J. Clinical Chemistry. Fourth edition, Mosby Press, UK, London, 2000.
10. Methods in Enzymology. Edited by Lester Packer, University of California, Academic Press,
USA, 1999.
11. Metzler D.E. Biochemistry. The chemical reactions of living cells. Second Edition, vol. 1-2.
Academic Press, 2003.
12. Murray R.K., Granner D.K., Mayes P.A., Rodwell V.W. Harpers illustrated Biochemistry.
Twenty-sixth International Edition, 2003.
13. Nelson D.L., Cox M.M. Lehninger principles of Biochemistry. 3-rd Edition p.cm. Includes
index, 2000.
14. Oral Bacterial Ecology: the Molecular Basis. Edited by Howard K. Kuramitsu (USA,
Buffalo) and Richard P. Ellen (Canada, Toronto). Horizon Scientific Press, UK, 2000.
15. Russel P.J. Genetics, 3
rd
Edition. Harper Collins Publischers, New York, 1992.
16. Stroev E.A. Biochemistry. Russia, Moscow, 1989.
17. Stryer L. Biochemistry. W.H.Freeman and Company, San Francisco, USA, 2000.



























138
CONTENTS

Introduction. 3
Lecture 1. Introduction in Biochemistry. Amino acids. 4
Lecture 2. Proteins: structure and levels of structural organizations of proteins.. 7
Lecture 3. Physical and chemical properties of proteins.. 11
Lecture 4. Enzymes: classification, nomenclature and chemical structure of enzymes.
Coenzymes.. 15
Lecture 5. Mechanism of enzymic action, regulation of enzyme activity 19
Lecture 6. Nucleic acids: structure and levels of nucleic acids organization.. 24
Lecture 7. Types of genetic transfer. Molecular fundamentals of replication and trans-
cription 30
Lecture 8. Molecular principles of translation.. 36
Lecture 9. General characterization of metabolism and energy metabolism. 41
Lecture 10. Enzymic mitochondrial systems as hydrogen generations... 47
Lecture 11. Carbohydrates: chemical structure, digestive mechanism of sugars.. 49
Lecture 12. Glycolysis anaerobic oxidation of carbohydrates.. 54
Lecture 13. Pentose phosphate cycle. Carbohydrate metabolism control in the human
organism.................................................................................................................. 60
Lecture 14. Lipids: classification, chemical structure and digestion of lipids.... 62
Lecture 15. Catabolism of lipids in tissues. 68
Lecture 16. Biosynthesis of lipids in tissues. Regulation and pathology of lipid metabolism 71
Lecture 17. Catabolism of simple proteins. 77
Lecture 18. Amino acid catabolic pathways in tissues.. 80
Lecture 19. Routes to metabolic detoxification of ammonia. 82
Lecture 20. Conversion of amino acids to specialized products... 85
Lecture 21. Interrelation of major metabolic routes in the organism. Metabolism of nuc-
leoproteins.... 89
Lecture 22. Hemoprotein metabolism..... 90
Lecture 23. Neuroendocrine control of metabolism.. 94
Lecture 24. Hormones of hypothalamus-hypophyseal system. Hormones of peripheral
glands... 98
Lecture 25. Biochemistry of blood: chemical composition of blood plasma....... 103
Lecture 26. Respiratory function of the blood. Hemostatic function of blood.... 106
Lecture 27. Functional biochemistry of the liver... 110
Lecture 28. Functional biochemistry of kidney. 112
Lecture 29. Biotransformation of drugs and poisons 114
Lecture 30. Biochemistry of connective tissue... 120
Lecture 31. Biochemistry of bone and teeth... 123
Lecture 32. Biochemistry of saliva.. 127
Lecture 33. Biochemistry of muscle tissue.. 129
Lecture 34. Production of energy in photosynthetic organisms... 132
Literature 136

You might also like